Skip to main content

Full text of "USPTO Patents Application 10551504"

See other formats


(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property 
Organization 

International Bureau 

(43) International Publication Date 
8 January 2004 (08.01.2004) 





PCT 



(10) International Publication Number 

WO 2004/003019 A2 



(51) International Patent Classification 7 : 



C07K 16/00 



(21) International Application Number: 

PCT/GB2003/002804 

(22) International Filing Date: 30 June 2003 (30.06.2003) 



(25) Filing Language: 

(26) Publication Language: 



English 
English 



(30) Priority Data: 

PCT/GB02/03014 28 June 2002 (28.06.2002) GB 

0230202.4 27 December 2002 (27.12.2002) GB 

(71) Applicant (for all designated States except US): DOMAN- 
TIS LIMITED [GB/GB]; Granta Park, Abington, Cam- 
bridgeshire CB1 6GS (GB). 

(72) Inventors; and 

(75) Inventors/Applicants (for US only): WINTER, Greg 
[GB/GB]; MRC Laboratory of Molecular Biology, Hills 
Road, Cambridge CB2 2QH (GB). TOMLINSON, Ian 
[GB/GB]; Domantis Limited, Granta Park, Abington, 
Cambridge CB1 6GS (GB). IGNATOVICH, Olga 
[BY/GB]; Domantis Limited, Granta Park, Abington, 
Cambridge CB1 6GS (GB). HOLT, Lucy [GB/GB]; 
Domantis Limited, Granta Park, Abington, Cambridge 
CB1 6GS (GB). DE ANGELIS, Elena [IT/GB]; Domantis 
Limited, Granta Park, Abington, Cambridge CB1 6GS 
(GB). 



(74) Agents: MASCHIO, Antonio et al.; D Young & Co, 21 
New Fetter Lane, London EC4A IDA (GB). 

(81) Designated States (national): AE, AG, AL, AM, AT, AU, 
AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CO, CR, CU, 
CZ, DE, DK, DM, DZ, EC, EE, ES, FI, GB, GD, GE, GH, 
GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, 
LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, 
MX, MZ, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, 
SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, 
UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. 

(84) Designated States (regional): ARIPO patent (GH, GM, 
KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZM, ZW), 
Eurasian patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), 
European patent (AT, BE, BG, CH, CY, CZ, DE, DK, EE, 
ES, FI, FR, GB, GR, HU, IE, IT, LU, MC, NL, PT, RO, 
SE, SI, SK, TR), OAPI patent (BF, BJ, CF, CG, CI, CM, 
GA, GN, GQ, GW, ML, MR, NE, SN, TD, TG). 

Declaration under Rule 4.17: 

— of inventorship ( Rule 4. 1 7( iv ) ) for US only 

Published: 

— without international search report and to be republished 
upon receipt of that report 

For two-letter codes and other abbreviations ; refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



< 



(54) Title: LIGAND 

O 



(57) Abstract: The invention provides a dual-specific ligand comprising a first immunoglobulin variable domain having a first bind- 
ing specificity and a complementary or non-complementary immunoglobulin variable domain having a second binding specificity. 



WO 2004/003019 



PCT/GB2003/002804 



Ligand 

The present invention relates to dual specific ligands. In particular, the invention 
provides a method for the preparation of dual-specific ligands comprising a first 
5 immunoglobulin single variable domain binding to a first antigen or epitope, and a second 
immunoglobulin single variable domain binding to a second antigen or epitope. More 
particularly, the invention relates to dual-specific ligands wherein binding to at least one 
of the first and second antigens or epitopes acts to increase the half-life of the ligand in 
vivo. Open and closed conformation ligands comprising more than one binding specificity 
10 are described. 

Introduction 

The antigen binding domain of an antibody comprises two separate regions: a heavy 

15 chain variable domain (v H ) anci a light chain variable domain (v L : which can be either 
V K or Vx). The antigen binding site itself is formed by six polypeptide loops: three from 
V H domain (HI, H2 and H3) and three from V L domain (LI, L2 and L3). A diverse 
primary repertoire of V genes that encode the v H and V L domains is produced by the 
combinatorial rearrangement of gene segments. The v H § ene is produced by the 

20 recombination of three gene segments, v H > D and J H- In humans, there are approximately 
51 functional V H segments (Cook and Tomlinson (1995) Immunol Today, 16: 237), 25 
functional D segments (Corbett et al (1997) J. Mol Biol, 268: 69) and 6 functional Jh 
segments (Ravetch et al. (1981) Cell, 27: 583), depending on the haplotype. The v H 
segment encodes the region of the polypeptide chain which forms the first and second 

25 antigen binding loops of the v H domain (HI and H2), whilst the y H , D and J H segments 
combine to form the third antigen binding loop of the y H domain (H3). The v L S ene is 
produced by the recombination of only two gene segments, v L and hi humans, there 
are approximately 40 functional V K segments (Schable and Zachau (1993) Biol Chem. 
Hoppe-Seyler, 374: 1001), 31 functional V% segments (Williams et al (1996) J. Mol 

30 Biol, 264: 220; Kawasaki et al (1997) Genome Res., 7: 250), 5 functional J K segments 
(Hieter et al (1982) J. Biol Chem., 257: 1516) and 4 functional segments (Vasicek 



WO 2004/003019 PCT/GB2003/002804 

2 

and Leder (1990) J. Exp. Med., 172: 609), depending on the haplotype. The v L segment 
encodes the region of the polypeptide chain which forms the first and second antigen 
binding loops of the y L domain (LI and L2), whilst the Vl an d J L segments combine to 
form the third antigen binding loop of the Vl domain (L3). Antibodies selected from this 
5 primary repertoire are believed to be sufficiently diverse to bind almost all antigens with 
at least moderate affinity. High affinity antibodies are produced by "affinity maturation" 
of the rearranged genes, in which point mutations are generated and selected by the 
immune system on the basis of improved binding. 

10 Analysis of the structures and sequences of antibodies has shown that five of the six 
antigen binding loops (HI, H2, LI, L2, L3) possess a limited number of main-chain 
conformations or canonical structures (Chothia and Lesk (1987) J. MoL Biol, 196: 901; 
Chothia et al (1989) Nature, 342: 877). The main-chain conformations are determined by 
(i) the length of the antigen binding loop, and (ii) particular residues, or types of residue, 

15 at certain key position in the antigen binding loop and the antibody framework. Analysis 
of the loop lengths and key residues has enabled us to the predict the main-chain 
conformations of HI, H2, LI, L2 and L3 encoded by the majority of human antibody 
sequences (Chothia et al (1992) J. MoL Biol, 227: 799; Tomlinson et al (1995) EMBO 
J., 14: 4628; Williams et al (1996) J. MoL Biol, 264: 220). Although the H3 region is 

20 much more diverse in terms of sequence, length and structure (due to the use of D 
segments), it also forms a limited number of main-chain conformations for short loop 
lengths which depend on the length and the presence of particular residues, or types of 
residue, at key positions in the loop and the antibody framework (Martin et al (1996) J. 
MoL Biol, 263: 800; Shirai et al (1996) FEBS Letters, 399: 1. 

25 

Bispecific antibodies comprising complementary pairs of Vh an d V L regions are known in 
the art. These bispecific antibodies must comprise two pairs of Vh an d V L S > ea °h WVl 
pair binding to a single antigen or epitope. Methods described involve hybrid hybridomas 
(Milstein & Cuello AC, Nature 305:537-40), minibodies (Hu et al, (1996) Cancer Res 
30 56:3055-3061;), diabodies (Holliger et al, (1993) Proc. Natl. Acad. Sci. USA 90, 6444- 
6448; WO 94/13804), chelating recombinant antibodies (CRAbs; (Neri et al, (1995) J. 
MoL Biol. 246, 367-373), biscFv (e.g. Atwell et al, (1996) Mol. Immunol. 33, 1301- 
1312), "knobs in holes" stabilised antibodies (Carter et al, (1997) Protein Sci. 6, 781- 



WO 2004/003019 PCT/GB2003/002804 

3 

788). In each case each antibody species comprises two antigen-binding sites, each 
fashioned by a complementary pair of V H and V L domains. Each antibody is thereby able 
to bind to two different antigens or epitopes at the same time, with the binding to EACH 
antigen or epitope mediated by a v H and its complementary v L domain. Each of these 

5 techniques presents its particular disadvantages; for instance in the case of hybrid 
hybridomas, inactive V n /y L pairs can greatly reduce the fraction of bispecific IgG. 
Furthermore, most bispecific approaches rely on the association of the different V H /V L 
pairs or the association of v H and V L chains to recreate the two different Vi/Vl binding 
sites. It is therefore impossible to control the ratio of binding sites to each antigen or 

10 epitope in the assembled molecule and thus many of the assembled molecules will bind 
to one antigen or epitope but not the other. In some cases it has been possible to engineer 
the heavy or light chains at the sub-unit interfaces (Carter et al 9 1997) in order to improve 
the number of molecules which have binding sites to both antigens or epitopes but this 
never results in all molecules having binding to both antigens or epitopes. 

15 

There is some evidence that two different antibody binding specificities might be 
incorporated into the same binding site, but these generally represent two or more 
specificities that correspond to structurally related antigens or epitopes or to antibodies 
that are broadly cross-reactive.. For example, cross-reactive antibodies have been 

20 described, usually where the two antigens are related in sequence and structure, such as 
hen egg white lysozyme and turkey lysozyme (McCafferty et al., WO 92/01047) or to 
free hapten and to hapten conjugated to carrier (Griffiths AD et al. EMBO J 1994 13:14 
3245-60). In a further example, WO 02/02773 (Abbott Laboratories) describes antibody 
molecules with "dual specificity". The antibody molecules referred to are antibodies 

25 raised or selected against multiple antigens, such that their specificity spans more than a 
single antigen. Each complementary Vi/Vl P air in the antibodies of WO 02/02773 
specifies a single binding specificity for two or more structurally related antigens; the v H 
and v L domains in such complementary pairs do not each possess a separate specificity. 
The antibodies thus have a broad single specificity which encompasses two antigens, 

30 which are structurally related. Furthermore natural autoantibodies have been described 
that are polyreactive (Casali & Notkins, Ann. Rev. Immunol. 7, 515-531), reacting with at 
least two (usually more) different antigens or epitopes that are not structurally related. It 



WO 2004/003019 PCT/GB2003/002804 

4 

has also been shown that selections of random peptide repertoires using phage display 
technology on a monoclonal antibody will identify a range of peptide sequences that fit 
the antigen binding site. Some of the sequences are highly related, fitting a consensus 
sequence, whereas others are very different and have been termed mimotopes (Lane & 
5 Stephen, Current Opinion in Immunology, 1993, 5, 268-271). It is therefore clear that a 
natural four-chain antibody, comprising associated and complementary v H and V L 
domains, has the potential to bind to many different antigens from a large universe of 
known antigens. It is less clear how to create a binding site to two given antigens in the 
same antibody, particularly those which are not necessarily structurally related. 

10 

Protein engineering methods have been suggested that may have a bearing on this. For 
example it has also been proposed that a catalytic antibody could be created with a 
binding activity to a metal ion through one variable domain, and to a hapten (substrate) 
through contacts with the metal ion and a complementary variable domain (Barb as et al., 
15 1993 Proc. Natl. Acad. Sci USA 90, 6385-6389). However in this case, the binding and 
catalysis of the substrate (first antigen) is proposed to require the binding of the metal ion 
(second antigen). Thus the binding to the Vj/Vl pairing relates to a single but multi- 
component antigen. 

20 Methods have been described for the creation of bispecific antibodies from camel 
antibody heavy chain single domains in which binding contacts for one antigen are 
created in one variable domain, and for a second antigen in a second variable domain. 
However the variable domains were not complementary. Thus a first heavy chain variable 
domain is selected against a first antigen, and a second heavy chain variable domain 

25 against a second antigen, and then both domains are linked together on the same chain to 
give a bispecific antibody fragment (Conrath et al., J. Biol. Chem. 270, 27589-27594). 
However the camel heavy chain single domains are unusual in that they are derived from 
natural camel antibodies which have no light chains, and indeed the heavy chain single 
domains are unable to associate with camel light chains to form complementary v H ^ d 

30 VLP a ^ rs * 

Single heavy chain variable domains have also been described, derived from natural 
antibodies which are normally associated with light chains (from monoclonal antibodies 



WO 2004/003019 PCT/GB2003/002804 

5 

or from repertoires of domains; see EP-A-0368684). These heavy chain variable 
domains have been shown to interact specifically with one or more related antigens but 
have not been combined with other heavy or light chain variable domains to create a 
ligand with a specificity for two or more different antigens . Furthermore, these single 
domains have been shown to have a very short in vivo half-life. Therefore such domains 
are of limited therapeutic value. 

It has been suggested to make bispecific antibody fragments by linking heavy chain 
variable domains of different specificity together (as described above). The disadvantage 
with this approach is that isolated antibody variable domains may have a hydrophobic 
interface that normally makes interactions with the light chain and is exposed to solvent 
and may be "sticky" allowing the single domain to bind to hydrophobic surfaces. 
Furthermore, in the absence of a partner light chain the combination of two or more 
different heavy chain variable domains and their association, possibly via their 
hydrophobic interfaces, may prevent them from binding to one in not both of the ligands 
they are able to bind in isolation. Moreover, in this case the heavy chain variable 
domains would not be associated with complementary light chain variable domains and 
thus may be less stable and readily unfold (Worn & Pluckthun, 1998 Biochemistry 37, 
13120-7). 

Summary of the invention 

The inventors have described, in their copending international patent application WO 
03/002609 as well as copending unpublished UK patent application 0230203.2, dual 
specific immunoglobulin ligands which comprise immunoglobulin single variable 
domains which each have different specificities. The domains may act in competition 
with each other or independently to bind antigens or epitopes on target molecules. 

In a first configuration, the present invention provides a further improvement in dual 
specific ligands as developed by the present inventors, in which one specificity of the 
ligand is directed towards a protein or polypeptide present in vivo in an organism which 
can act to increase the half-life of the ligand by binding to it. 



WO 2004/003019 PCT/GB2003/002804 

6 

Accordingly, in a first aspect, there is provided a dual-specific ligand comprising a first 
immunoglobulin single variable domain having a binding specificity to a first antigen or 
epitope and a second complementary immunoglobulin single variable domain having a 
binding activity to a second antigen or epitope, wherein one or both of said antigens or 
epitopes acts to increase the half-life of the ligand in vivo and wherein said first and 
second domains lack mutually complementary domains which share the same specificity, 
provided that said dual specific ligand does not consist of an anti-HSA V H domain and an 
anti-P galactosidase V K domain. Preferably, that neither of the first or second variable 
domains binds to human serum albumin (HSA). 

Antigens or epitopes which increase the half-life of a ligand as described herein are 
advantageously present on proteins or polypeptides found in an organism in vivo. 
Examples include extracellular matrix proteins, blood proteins, and proteins present in 
various tissues in the organism. The proteins act to reduce the rate of ligand clearance 
from the blood, for example by acting as bulking agents, or by anchoring the ligand to a 
desired site of action. Examples of antigens/epitopes which increase half-life in vivo are 
given in Annex 1 below. 

Increased half-life is useful in in vivo applications of immuno globulins, especially 
antibodies and most especially antibody fragments of small size. Such fragments (Fvs, 
disulphide bonded Fvs, Fabs, scFvs, dAbs) suffer from rapid clearance from the body; 
thus, whilst they are able to reach most parts of the body rapidly, and are quick to produce 
and easier to handle, their in vivo applications have been limited by their only brief 
persistence in vivo. The invention solves this problem by providing increased half-life of 
the ligands in vivo and consequently longer persistence times in the body of the functional 
activity of the ligand. 

Methods for pharmacokinetic analysis and determination of ligand half-life will be 
familiar to those skilled in the art. Details may be found in Kenneth, A et al: Chemical 
Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al 
Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to 
"Pharmacokinetics", M Gibaldi & D Perron, published by Marcel Dekker, 2 nd Rev. ex 



WO 2004/003019 PCT/GB2003/002804 

7 

edition (1982), which describes pharmacokinetic parameters such as t alpha and t beta 
half lives and area under the curve (AUC). 

Half lives (tV 2 alpha and t l A beta) and AUC can be determined from a curve of serum 
concentration of ligand against time. The WinNonlin analysis package (available from 
Pharsight Corp., Mountain View, CA94040, USA) can be used, for example, to model the 
curve. In a first phase (the alpha phase) the ligand is undergoing mainly distribution in 
the patient, with some elimination. A second phase (beta phase) is the terminal phase 
when the ligand has been distributed and the serum concentration is decreasing as the 
ligand is cleared from the patient. The t alpha half life is the half life of the first phase 
and the t beta half life is the half life of the second phase. Thus, advantageously, the 
present invention provides a ligand or a composition comprising a ligand according to the 
invention having a ta half-life in the range of 15 minutes or more. In one embodiment, 
the lower end of the range is 30 minutes, 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 
hours, 6 hours, 7 hours, 10 hours, 11 hours or 12 hours. In addition, or alternatively^ 
ligand or composition according to the invention will have a ta half life in the range of up 
to and including 12 hours. In one embodiment, the upper end of the range is 11, 10, 9, 8, 
7, 6 or 5 hours. An example of a suitable range is 1 to 6 hours, 2 to 5 hours or 3 to 4 
hours. 

Advantageously, the present invention provides a ligand or a composition comprising a 
ligand according to the invention having a tp half-life in the range of 2.5 hours or more. 
In one embodiment, the lower end of the range is 3 hours, 4 hours, 5 hours, 6 hours, 7 
hours, 10 hours , 11 hours, or 12 hours. In addition, or alternatively, a ligand or 
composition according to the invention has a tp half-life in the range of up to and 
including 21 days. In one embodiment, the upper end of the range is 12 hours, 24 hours, 
2 days, 3 days, 5 days, 10 days, 15 days or 20 days. Advantageously a ligand or 
composition according to the invention will have a tp half life in the range 12 to 60 hours. 
In a further embodiment, it will be in the range 12 to 48 hours. In a further embodiment 
still, it will be in the range 12 to 26 hours. 



In addition, or alternatively to the above criteria, the present invention provides a ligand 
or a composition comprising a ligand according to the invention having an AUC 



WO 2004/003019 PCT/GB2003/002804 

8 

value (area under the curve) in the range of 1 mg.min/ml or more. In one embodiment, 
the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300mg.min/ml. In addition, or 
alternatively, a ligand or composition according to the invention has an AUC in the range 
of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 
5 200, 150, 100, 75 or 50 mg.min/ml. Advantageously a ligand according to the invention 
will have a AUC in the range selected from the group consisting of the following: 1 5 to 
150mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50mg.min/ml. 

In a first embodiment, the dual specific ligand comprises two complementary variable 
10 domains, i.e. two variable domains that, in their natural environment, are capable of 
operating together as a cognate pair or group even if in the context of the present 
invention they bind separately to their cognate epitopes. For example, the complementary 
variable domains may be immunoglobulin heavy chain and light chain variable domains 
(V H and V L ). V H and V L domains are advantageously provided by scFv or Fab antibody 
15 fragments. Variable domains may be linked together to form multivalent ligands by, for 
example: provision of a hinge region at the C-terminus of each V domain and disulphide 
bonding between cysteines in the hinge regions; or provision of dAbs each with a cysteine 
at the C-terminus of the domain, the cysteines being disulphide bonded together; or 
production of V-CH & V-CL to produce a Fab format; or use of peptide linkers (for 
20 example Gly 4 Ser linkers discussed hereinbelow) to produce dimers, trimers and further 
multimers. 

The inventors have found that the use of complementary variable domains allows the two 
domain surfaces to pack together and be sequestered from the solvent. Furthermore the 

25 complementary domains are able to stabilise each other. In addition, it allows the creation 
of dual-specific IgG antibodies without the disadvantages of hybrid hybridomas as used 
in the prior art, or the need to engineer heavy or light chains at the sub-unit interfaces. 
The dual-specific ligands of the first aspect of the present invention have at least one 
V H /V L pair. A bispecific IgG according to this invention will therefore comprise two 

30 such pairs, one pair on each arm of the Y-shaped molecule. Unlike conventional 
bispecific antibodies or diabodies, therefore, where the ratio of chains used is 
determinative in the success of the preparation thereof and leads to practical difficulties, 
the dual specific ligands of the invention are free from issues of chain balance. Chain 



WO 2004/003019 PCT/GB2003/002804 

9 

imbalance in conventional bi-specific antibodies results from the association of two 
different V L chains with two different V H chains, where V L chain 1 together with V H 
chain 1 is able to bind to antigen or epitope 1 and V L chain 2 together with V H chain 2 is 
able to bind to antigen or epitope 2 and the two correct pairings are in some way linked to 
5 one another. Thus, only when V L chain 1 is paired with V H chain 1 and V L chain 2 is 
paired with V H chain 2 in a single molecule is bi-specificity created. Such bi-specific 
molecules can be created in two different ways. Firstly, they can be created by association 
of two existing V H /V L pairings that each bind to a different antigen or epitope (for 
example, in a bi-specific IgG). In this case the Vh/V l pairings must come all together in a 

10 1:1 ratio in order to create a population of molecules all of which are bi-specific. This 
never occurs (even when complementary CH domain is enhanced by "knobs into holes" 
engineering) leading to a mixture of bi-specific molecules and molecules that are only 
able to bind to one antigen or epitope but not the other. The second way of creating a bi- 
specific antibody is by the simultaneous association of two different Vh chain with two 

15 different V L chains (for example in a bi-specific diabody). In this case, although there 
tends to be a preference for Vl chain 1 to pair with V H chain 1 and V L chain 2 to pair with 
V H chain 2 (which can be enhanced by "knobs into holes" engineering of the V L and V H 
domains), this paring is never achieved in all molecules, leading to a mixed formulation 
whereby incorrect pairings occur that are unable to bind to either antigen or epitope. 

20 

Bi-specific antibodies constructed according to the dual-specific ligand approach 
according to the first aspect of the present invention overcome all of these problems 
because the binding to antigen or epitope 1 resides within the Vh or Vl domain and the 
binding to antigen or epitope 2 resides with the complementary V L or V H domain, 
25 respectively. Since V H and V L domains pair on a 1:1 basis all V H /V L pairings will be bi- 
specific and thus all formats constructed using these V H /V L pairings (Fv, scFvs, Fabs, 
minibodies, IgGs etc) will have 100% bi-specific activity. 

In the context of the present invention, first and second "epitopes" are understood to be 
30 epitopes which are not the same and are not bound by a single monospecific ligand. In 
the first configuration of the invention, they are advantageously on different antigens, one 
of which acts to increase the half-life of the ligand in vivo. Likewise, the first and second 
antigens are advantageously not the same. 



WO 2004/003019 



10 



PCT/GB2003/002804 



The dual specific ligands of the invention do not include ligands as described in WO 
02/02773. Thus, the ligands of the present invention do not comprise complementary 
Vh/V l pairs which bind any one or more antigens or epitopes co-operatively. Instead, the 
5 ligands according to the first aspect of the invention comprise a Vh/V l complementary 
pair, wherein the V domains have different specificities. 

Moreover, the ligands according to the first aspect of the invention comprise V h /Vl 
complementary pairs having different specificities for non-structurally related epitopes or 
10 antigens. Structurally related epitopes or antigens are epitopes or antigens which possess 
sufficient structural similarity to be bound by a conventional V H /V L complementary pair 
which acts in a co-operative manner to bind an antigen or epitope; in the case of 
structurally related epitopes, the epitopes are sufficiently similar in structure that they 
"fit" into the same binding pocket formed at the antigen binding site of the V H /V L dimer. 

15 

In a second aspect, the present invention provides a ligand comprising a first 
immunoglobulin variable domain having a first antigen or epitope binding specificity and 
a second immunoglobulin variable domain having a second antigen or epitope binding 
specificity wherein one or both of said first and second variable domains bind to an 
20 antigen which increases the half-life of the ligand in vivo, and the variable domains are 
not complementary to one another. 

In one embodiment, binding to one variable domain modulates the binding of the ligand 
to the second variable domain. 

25 

In this embodiment, the variable domains may be, for example, pairs of V H domains or 
pairs of Vl domains. Binding of antigen at the first site may modulate, such as enhance or 
inhibit, binding of an antigen at the second site. For example, binding at the first site at 
least partially inhibits binding of an antigen at a second site. In such an embodiment, the 
30 ligand may for example be maintained in the body of a subject organism in vivo through 
binding to a protein which increases the half-life of the ligand until such a time as it 
becomes bound to the second target antigen and dissociates from the half-life increasing 
protein. 



WO 2004/003019 



11 



PCT/GB2003/002804 



Modulation of binding in the above context is achieved as a consequence of the structural 
proximity of the antigen binding sites relative to one another. Such structural proximity 
can be achieved by the nature of the structural components linking the two or more 
5 antigen binding sites, eg by the provision of a ligand with a relatively rigid structure that 
holds the antigen binding sites in close proximity. Advantageously, the two or more 
antigen binding sites are in physically close proximity to one another such that one site 
modulates the binding of antigen at another site by a process which involves steric 
hindrance and/or conformational changes within the immunoglobulin molecule. 

10 

The first and the second antigen binding domains may be associated either covalently or 
non-covalently. In the case that the domains are covalently associated, then the 
association may be mediated for example by disulphide bonds or by a polypeptide linker 
such as (Gly4Ser) n? where n = from 1 to 8, eg, 2, 3, 4, 5 or 7. 

15 

Ligands according to the invention may be combined into non-immuno globulin multi- 
ligand structures to form multivalent complexes, which bind target molecules with the 
same antigen, thereby providing superior avidity, while at least one variable domain binds 
an antigen to increase the half life of the multimer. For example natural bacterial 

20 receptors such as SpA have been used as scaffolds for the grafting of CDRs to generate 
ligands which bind specifically to one or more epitopes. Details of this procedure are 
described in US 5,831,012. Other suitable scaffolds include those based on fibronectin 
and affibodies. Details of suitable procedures are described in WO 98/58965. Other 
suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. 

25 Mol. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 
(Medical Research Council), which are based for example on the ring structure of 
bacterial GroEL or other chaperone polypeptides. 

Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 
30 scaffold and used together with immunoglobulin Vh or V L domains to form a ligand. 
Likewise, fibronectin, lipocallin and other scaffolds maybe combined. 



WO 2004/003019 PCT/GB2003/002804 

12 

In the case that the variable domains are selected from V-gene repertoires selected for 
instance using phage display technology as herein described, then these variable domains 
can comprise a universal framework region, such that is they may be recognised by a 
specific generic ligand as herein defined. The use of universal frameworks, generic 
5 ligands and the like is described in WO99/20749. In the present invention, reference to 
phage display includes the use of both phage and/or phagemids. 

/■ 

Where V-gene repertoires are used variation in polypeptide sequence is preferably located 
within the structural loops of the variable domains. The polypeptide sequences of either 
10 variable domain may be altered by DNA shuffling or by mutation in order to enhance the 
interaction of each variable domain with its complementary pair. 

In a preferred embodiment of the invention the 'dual-specific ligand 5 is a single chain Fv 
fragment. In an alternative embodiment of the invention, the ' dual- specific ligand 5 
15 consists of a Fab region of an antibody. The term "Fab region 55 includes a Fab-like 
region where two VH or two VL domains are used. 

The variable regions may be derived from antibodies directed against target antigens or 
epitopes. Alternatively they may be derived from a repertoire of single antibody domains 
20 such as those expressed on the surface of filamentous bacteriophage. Selection may be 
performed as described below. 

In a third aspect, the invention provides a method for producing a ligand comprising a 
first immunoglobulin single variable domain having a first binding specificity and a 
25 second single immunoglobulin single variable domain having a second (different) binding 
specificity, one or both of the binding specificities being specific for an antigen which 
increases the half-life of the ligand in vivo, the method comprising the steps of: 

(a) selecting a first variable domain by its ability to bind to a first epitope, 

(b) selecting a second variable region by its ability to bind to a second epitope, 
30 (c) combining the variable domains; and 

(d) selecting the ligand by its ability to bind to said first epitope and to said second 
epitope. 



WO 2004/003019 PCT/GB2003/002804 

13 

The ligand can bind to the first and second epitopes either simultaneously or, where there 
is competition between the binding domains for epitope binding, the binding of one 
domain may preclude the binding of another domain to its cognate epitope. In one 
embodiment, therefore, step (d) above requires simultaneous binding to both first and 
5 second (and possibly further) epitopes; in another embodiment, the binding to the first 
and second epitoes is not simultaneous. 

The epitopes are preferably on separate antigens. 

10 Ligands advantageously comprise V h /Vl combinations, or V h /Vh or Vi/Vl combinations 
of immunoglobulin variable domains, as described above. The ligands may moreover 
comprise camelid Vhh domains, provided that the Vhh domain which is specific for an 
antigen which increases the half-life of the ligand in vivo does not bind Hen egg white 
lysozyme (HEL), porcine pancreatic alpha-amylase or NmC-A; hcg, BSA-linked RR6 azo 

15 dye or S. mutans HG982 cells, as described in Conrath et ah, (2001) JBC 276:7346-7350 
and W099/23221, neither of which describe the use of a specificity for an antigen which 
increases half-life to increase the half life of the ligand in vivo. 

In one embodiment, said first variable domain is selected for binding to said first epitope 
20 in absence of a complementary variable domain. In a further embodiment, said first 
variable domain is selected for binding to said first epitope/antigen in the presence of a 
third variable domain in which said third variable domain is different from said second 
variable domain and is complementary to the first domain. Similarly, the second domain 
may be selected in the absence or presence of a complementary variable domain. 

25 

The antigens or epitopes targeted by the ligands of the invention, in addition to the half- 
life enhancing protein, may be any antigen or epitope but advantageously is an antigen or 
epitope that is targeted with therapeutic benefit. The invention provides ligands, 
including open conformation, closed conformation and isolated dAb monomer ligands, 
30 specific for any such target, particularly those targets further identified herein. Such 
targets may be, or be part of, polypeptides, proteins or nucleic acids, which may be 
naturally occurring or synthetic. In this respect, the ligand of the invention may bind the 
epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist). One 



WO 2004/003019 PCT/GB2003/002804 

14 

skilled in the art will appreciate that the choice is large and varied. They may be for 
instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for 
enzymes or DNA binding proteins. Suitable cytokines and growth factors include but are 
not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin- 1 , EGF, EGF receptor, ENA-78, 
5 Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, 
FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF-pl, insulin, IFN-y, 
IGF-I, IGF-II, IL-la, IL-lp, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 
a.a.), IL-9, IL-10, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Inhibin a, 
Inhibin P, IP- 10, keratinocyte growth factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, 

10 Mullerian inhibitory substance, monocyte colony inhibitory factor, monocyte attractant 
protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-1 (MCAF), MCP-2, MCP-3, MCP- 
4, MDC (67 a.a.), MDC (69 a.a.), MIG, MIP-lcx, MIP-lp, MIP-3a, MIP-3p, MIP-4, 
myeloid progenitor inhibitor factor- 1 (MPIF-1), NAP -2, Neurturin, Nerve growth factor, 
p-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, 

15 SDFla, SDFlp, SCF, SCGF, stem cell factor (SCF), TARC, TGF-a, TGF-P, TGF-p2, 
TGF-P3, tumour necrosis factor (TNF), TNF-a, TNF-p, TNF receptor I, TNF receptor II, 
TNIL-1, TPO, VEGF, VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, 
GRO/MGSA, GRO-p, GRO-y, HCC1, 1-309, HER 1, HER 2, HER 3 and HER 4. 
Cytokine receptors include receptors for the foregoing cytokines. It will be appreciated 

20 that this list is by no means exhaustive. 

In one embodiment of the invention, the variable domains are derived from a respective 
antibody directed against the antigen or epitope. In a preferred embodiment the variable 
domains are derived from a repertoire of single variable antibody domains. 

25 

In a further aspect, the present invention provides one or more nucleic acid molecules 
encoding at least a dual-specific ligand as herein defined. The dual specific ligand may 
be encoded on a single nucleic acid molecule; alternatively, each domain may be encoded 
by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid 
30 molecule, the domains may be expressed as a fusion polypeptide, in the manner of a scFv 
molecule, or may be separately expressed and subsequently linked together, for example 
using chemical linking agents. Ligands expressed from separate nucleic acids will be 
linked together by appropriate means. 



WO 2004/003019 



15 



PCT/GB2003/002804 



The nucleic acid may further encode a signal sequence for export of the polypeptides 
from a host cell upon expression and may be fused with a surface component of a 
filamentous bacteriophage particle (or other component of a selection display system) 
5 upon expression. 

In a further aspect the present invention provides a vector comprising nucleic acid 
encoding a dual specific ligand according to the present invention. 

10 In a yet further aspect, the present invention provides a host cell transfected with a vector 
encoding a dual specific ligand according to the present invention. 

Expression from such a vector may be configured to produce, for example on the surface 
of a bacteriophage particle, variable domains for selection. This allows selection of 
15 displayed variable regions and thus selection of 'dual-specific ligands 5 using the method 
of the present invention. 

The present invention further provides a kit comprising at least a dual-specific ligand 
according to the present invention. 

20 

Dual-Specific ligands according to the present invention preferably comprise 
combinations of heavy and light chain domains. For example, the dual specific ligand 
may comprise a Vh domain and a Vl domain, which may be linked together in the form 
of an scFv. In addition, the ligands may comprise one or more Ch or Cl domains. For 

25 example, the ligands may comprise a ChI domain, C H 2 or Ch3 domain, and/or a C L 
domain, Cpl, Cjli2, Cp3 or C|J,4 domains, or any combination thereof. A hinge region 
domain may also be included. Such combinations of domains may, for example, mimic 
natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or 
F(ab') 2 molecules. Other structures, such as a single arm of an IgG molecule comprising 

30 V H , V L , C h 1 and Cl domains, are envisaged. 

In a preferred embodiment of the invention, the variable regions are selected from single 
domain V gene repertoires. Generally the repertoire of single antibody domains is 



WO 2004/003019 PCT/GB2003/002804 

16 

displayed on the surface of filamentous bacteriophage. In a preferred embodiment each 
single antibody domain is selected by binding of a phage repertoire to antigen. 

In a preferred embodiment of the invention each single variable domain may be selected 
5 for binding to its target antigen or epitope in the absence of a complementary variable 
region. In an alternative embodiment, the single variable domains may be selected for 
binding to its target antigen or epitope in the presence of a complementary variable 
region. Thus the first single variable domain may be selected in the presence of a third 
complementary variable domain, and the second variable domain may be selected in the 
10 presence of a fourth complementary variable domain. The complementary third or fourth 
variable domain may be the natural cognate variable domain having the same specificity 
as the single domain being tested, or a non-cognate complementary domain — such as a 
"dummy" variable domain. 

15 Preferably, the dual specific ligand of the invention comprises only two variable domains 
although several such ligands may be incorporated together into the same protein, for 
example two such ligands can be incorporated into an IgG or a multimeric 
immunoglobulin, such as IgM. Alternatively, in another embodiment a plurality of dual 
specific ligands are combined to form a multimer. For example, two different dual 

20 specific ligands are combined to create a tetra-specific molecule. 

It will be appreciated by one skilled in the art that the light and heavy variable regions of 
a dual-specific ligand produced according to the method of the present invention may be 
on the same polypeptide chain, or alternatively, on different polypeptide chains. In the 
25 case that the variable regions are on different polypeptide chains, then they may be linked 
via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking 
group, or any other method known in the art. 



In a further aspect, the present invention provides a composition comprising a dual- 
30 specific ligand, obtainable by a method of the present invention, and a pharmaceutically 
acceptable carrier, diluent or excipient. 



WO 2004/003019 PCT/GB2003/002804 

17 

Moreover, the present invention provides a method for the treatment and/or prevention 
of disease using a c dual-specific ligand' or a composition according to the present 
invention. 

5 In a second configuration, the present invention provides multispecific ligands which 
comprise at least two non-complementary variable domains. For example, the ligands 
may comprise a pair of V H domains or a pair of V L domains. Advantageously, the 
domains are of non-camelid origin; preferably they are human domains or comprise 
human framework regions (FWs) and one or more heterologous CDRs. CDRs and 
10 framework regions are those regions of an immunoglobulin variable domain as defined in 
the Kabat database of Sequences of Proteins of Immunological Interest. 

Preferred human framework regions are those encoded by germline gene segments DP47 
and DPK9. Advantageously, FW1, FW2 and FWS of a V H or V L domain have the 
15 sequence of FW1, FW2 or FWS from DP47 or DPK9. The human frameworks may 
optionally contain mutations, for example up to about 5 amino acid changes or up to 
about 10 amino acid changes collectively in the human frameworks used in the ligands of 
the invention. 

20 The variable domains in the multispecific ligands according to the second configuration 
of the invention may be arranged in an open or a closed conformation; that is, they may 
be arranged such that the variable domains can bind their cognate ligands independently 
and simultaneously, or such that only one of the variable domains may bind its cognate 
ligand at any one time. 

25 

The inventors have realised that under certain structural conditions, non-complementary 
variable domains (for example two light chain variable domains or two heavy chain 
variable domains) may be present in a ligand such that binding of a first epitope to a first 
variable domain inhibits the binding of a second epitope to a second variable domain, 
30 even though such non-complementary domains do not operate together as a cognate pair. 



WO 2004/003019 PCT/GB2003/002804 

18 

Advantageously, the ligand comprises two or more pairs of variable domains; that is, it 
comprises at least four variable domains. Advantageously, the four variable domains 
comprise frameworks of human origin. 

5 In a preferred embodiment, the human frameworks are identical to those of human 
germline sequences. 

The present inventors consider that such antibodies will be of particular use in ligand 
binding assays for therapeutic and other uses. 

10 

Thus, in a first aspect of the second configuration, the present invention provides a 
method for producing a multispecific ligand comprising the steps of: 

a) selecting a first epitope binding domain by its ability to bind to a first epitope, 

b) selecting a second epitope binding domain by its ability to bind to a second 
15 epitope, 

c) combining the epitope binding domains; and 

d) selecting the closed conformation multispecific ligand by its ability to bind to said 
first second epitope and said second epitope. 



20 In a further aspect of the second configuration, the invention provides method for 
preparing a closed conformation multi-specific ligand comprising a first epitope binding 
domain having a first epitope binding specificity and a non-complementary second 
epitope binding domain having a second epitope binding specificity, wherein the first and 
second binding specificities compete for epitope binding such that the closed 

25 conformation multi-specific ligand may not bind both epitopes simultaneously, said 
method comprising the steps of: 



a) selecting a first epitope binding domain by its ability to bind to a first epitope, 

b) selecting a second epitope binding domain by its ability to bind to a second 
30 epitope, 

c) combining the epitope binding domains such that the domains are in a closed 
conformation; and 



WO 2004/003019 PCT/GB2003/002804 

19 

d) selecting the closed conformation multispecific ligand by its ability to bind to 
said first second epitope and said second epitope, but not to both said first and 
second epitopes simultaneously. 

5 Moreover, the invention provides a closed conformation multi-specific ligand comprising 
a first epitope binding domain having a first epitope binding specificity and a non- 
complementary second epitope binding domain having a second epitope binding 
specificity, wherein the first and second binding specificities compete for epitope binding 
such that the closed conformation multi-specific ligand may not bind both epitopes 
10 simultaneously. 

An alternative embodiment of the above aspect of the of the second configuration of the 
invention optionally comprises a further step (bl) comprising selecting a third or further 
epitope binding domain. In this way the multi-specific ligand produced, whether of open 
15 or closed conformation, comprises more than two epitope binding specificities. In a 
preferred aspect of the second configuration of the invention, where the multi-specific 
ligand comprises more than two epitope binding domains, at least two of said domains are 
in a closed conformation and compete for binding; other domains may compete for 
binding or may be free to associate independently with their cognate epitope(s). 

20 

According to the present invention the term c multi-specific ligand' refers to a ligand 
which possesses more than one epitope binding specificity as herein defined. 

As herein defined the term 'closed conformation' (multi-specific ligand) means that the 
25 epitope binding domains of the ligand are attached to or associated with each other, 
optionally by means of a protein skeleton, such that epitope binding by one epitope 
binding domain competes with epitope binding by another epitope binding domain. That 
is, cognate epitopes may be bound by each epitope binding domain individually but not 
simultaneosuly. The closed conformation of the ligand can be achieved using methods 
30 herein described. 

"Open conformation" means that the epitope binding domains of the ligand are attached 
to or associated with each other, optionally by means of a protein skeleton, such that 



WO 2004/003019 PCT/GB2003/002804 

20 

epitope binding by one epitope binding domain does not compete with epitope binding 
by another epitope binding domain. 

As referred to herein, the term 'competes' means that the binding of a first epitope to its 
5 cognate epitope binding domain is inhibited when a second epitope is bound to its 
cognate epitope binding domain. For example, binding may be inhibited sterically, for 
example by physical blocking of a binding domain or by alteration of the structure or 
environment of a binding domain such that its affinity or avidity for an epitope is reduced. 

10 hi a further embodiment of the second configuration of the invention, the epitopes may 
displace each other on binding. For example, a first epitope may be present on an antigen 
which, on binding to its cognate first binding domain, causes steric hindrance of a second 
binding domain, or a coformational change therein, which displaces the epitope bound to 
the second binding domain. 

15 

Advantageously, binding is reduced by 25% or more, advantageously 40%, 50%, 60%, 
70%, 80%, 90% or more, and preferably up to 100% or nearly so, such that binding is 
completely inhibited. Binding of epitopes can be measured by conventional antigen 
binding assays, such as ELISA, by fluorescence based techniques, including FRET, or by 
20 techniques such as suface plasmon resonance which measure the mass of molecules. 

According to the method of the present invention, advantageously, each epitope binding 
domain is of a different epitope binding specificity. 

25 In the context of the present invention, first and second "epitopes" are understood to be 
epitopes which are not the same and are not bound by a single monospecific ligand. They 
may be on different antigens or on the same antigen, but separated by a sufficient distance 
that they do not form a single entity that could be bound by a single mono-specific V H /V L 
binding pair of a conventional antibody. Experimentally, if both of the individual 

30 variable domains in single chain antibody form (domain antibodies or dAbs) are 
separately competed by a monospecific V H /V L ligand against two epitopes then those two 
epitopes are not sufficiently far apart to be considered separate epitopes according to the 
present invention. 



WO 2004/003019 



PCT/GB2003/002804 



21 



The closed conformation multispecific ligands of the invention do not include ligands as 
described in WO 02/02773. Thus, the ligands of the present invention do not comprise 
complementary V H /y L pairs which bind any one or more antigens or epitopes co- 
5 operatively. Instead, the ligands according to the invention preferably comprise non- 
complementary V H -V H or V L -V L P airs - Advantageously, each v H or V L domain in each 
V H -V H or V L -V L P air has a different epitope binding specificity, and the epitope binding 
sites are so arranged that the binding of an epitope at one site competes with the binding 
of an epitope at another site. 

10 

According to the present invention, advantageously, each epitope binding domain 
comprises an immunoglobulin variable domain. More advantageously, each 
immunoglobulin variable domain will be either a variable light chain domain (vj or a 
variable heavy chain domain V H - 111 the secon d configuration of the present invention, 

15 the immunoglobulin domains when present on a ligand according to the present 
invention are non-complementary, that is they do not associate to form a V n /V L antigen 
binding site. Thus, multi-specific ligands as defined in the second configuration of the 
invention comprise immunoglobulin domains of the same sub-type, that is either variable 
light chain domains (Vl) or variable heavy chain domains (v H )- Moreover, where the 

20 ligand according to the invention is in the closed conformation, the immunoglobulin 
domains may be of the camelid Vhh type. 

In an alternative embodiment, the ligand(s) according to the invention do not comprise a 
camelid V H h domain. More particularly, the ligand(s) of the invention do not comprise 
25 one or more amino acid residues that are specific to camelid V H h domains as compared to 
human Vh domains. 

Advantageously, the single variable domains are derived from antibodies selected for 
binding activity against different antigens or epitopes. For example, the variable domains 
30 may be isolated at least in part by human immunisation. Alternative methods are known 
in the art, including isolation from human antibody libraries and synthesis of artificial 
antibody genes. 



WO 2004/003019 PCT/GB2003/002804 

22 

The variable domains advantageously bind super antigens, such as protein A or protein L. 
Binding to superantigens is a property of correctly folded antibody variable domains, and 
allows such domains to be isolated from, for example, libraries of recombinant or mutant 
domains. 

Epitope binding domains according to the present invention comprise a protein scaffold 
and epitope interaction sites (which are advantageously on the surface of the protein 
scaffold). 

Epitope binding domains may also be based on protein scaffolds or skeletons other than 
immunoglobulin domains. For example natural bacterial receptors such as SpA have been 
used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to 
one or more epitopes. Details of this procedure are described in US 5,831,012. Other 
suitable scaffolds include those based on fibronectin and affibodies. Details of suitable 
procedures are described in WO 98/58965. Other suitable scaffolds include lipocallin and 
CTLA4, as described in van den Beuken et al 9 J. Mol. Biol. (2001) 310, 591-601, and 
scaffolds such as those described in WO0069907 (Medical Research Council), which are 
based for example on the ring structure of bacterial GroEL or other chaperone 
polypeptides. 

Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 
scaffold and used together with immunoglobulin V H or V L domains to form a multivalent 
ligand. Likewise, fibronectin, lipocallin and other scaffolds maybe combined. 

25 It will be appreciated by one skilled in the art that the epitope binding domains of a closed 
conformation multispecific ligand produced according to the method of the present 
invention may be on the same polypeptide chain, or alternatively, on different polypeptide 
chains. In the case that the variable regions are on different polypeptide chains, then they 
may be linked via a linker, advantageously a flexible linker (such as a polypeptide chain), 

30 a chemical linking group, or any other method known in the art. 



10 



15 



20 



WO 2004/003019 PCT/GB2003/002804 

23 

The first and the second epitope binding domains may be associated either covalently or 
non-covalently. In the case that the domains are covalently associated, then the 
association may be mediated for example by disulphide bonds. 

5 In the second configuation of the invention, the first and the second epitopes are 
preferably different. They may be, or be part of, polypeptides, proteins or nucleic acids, 
which may be naturally occurring or synthetic. In this respect, the ligand of the invention 
may bind an epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor 
agonist). The epitope binding domains of the ligand in one embodiment have the same 

10 epitope specificity, and may for example simultaneously bind their epitope when multiple 
copies of the epitope are present on the same antigen. In another embodiment, these 
epitopes are provided on different antigens such that the ligand can bind the epitopes and 
bridge the antigens. One skilled in the art will appreciate that the choice of epitopes and 
antigens is large and varied. They may be for instance human or animal proteins, 

15 cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins. 
Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, 
BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, 
EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine 
(CX3C), GDNF, G-CSF, GM-CSF, GF-pl, insulin, IFN-y, IGF-I, IGF-H, IL-la, IL-ip, 

20 IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL-12, 
IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Inhibin a, Inhibin p, IP-10, keratinocyte growth 
factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, Mullerian inhibitory substance, 
monocyte colony inhibitory factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), 
MDC (69 a.a.), MCP-1 (MCAF), MCP-2, MCP-3, MCP-4, MDC (67 a.a.), MDC (69 

25 a.a.), MIG, MlP-la, MIP-lp, MIP-3a, MIP-3P, MIP-4, myeloid progenitor inhibitor 
factor-1 (MPIF-1), NAP-2, Neurturin, Nerve growth factor, p-NGF, NT-3, NT-4, 
Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFla, SDFip, SCF, 
SCGF, stem cell factor (SCF), TARC, TGF-a, TGF-p, TGF-p2, TGF-p3, tumour necrosis 
factor (TNF), TNF-a, TNF-p, TNF receptor I, TNF receptor II, TNIL-1, TPO, VEGF, 

30 VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, GRO/MGSA, GRO-p, 
GRO-y, HCC1, 1-309, HER 1, HER 2, HER 3, HER 4, TACE recognition site, TNF BP-I 
and TNF BP-II, as well as any target disclosed in Annex 2 or Annex 3 hereto, whether in 
combination as set forth in the Annexes, in a different combination or individually. 



WO 2004/003019 PCT/GB2003/002804 

24 

Cytokine receptors include receptors for the foregoing cytokines, e.g. IL-1 Rl; IL-6R; 
IL-10R; IL-18R, as well as receptors for cytokines set forth in Annex 2 or Annex 3 and 
also receptors disclosed in Annex 2 and 3. It will be appreciated that this list is by no 
means exhaustive. Where the multispecific ligand binds to two epitopes (on the same or 
5 different antigens), the antigen(s) may be selected from this list. 

Advantageously, dual specific ligands may be used to target cytokines and other 
molecules which cooperate synergistically in therapeutic situations in the body of an 
organism. The invention therefore provides a method for synergising the activity of two 

10 or more cytokines, comprising administering a dual specific ligand capable of binding to 
said two or more cytokines. In this aspect of the invention, the dual specific ligand may 
be any dual specific ligand, including a ligand composed of complementary and/or non- 
complementary domains, a ligand in an open conformation, and a ligand in a closed 
conformation. For example, this aspect of the invention relates to combinations of V H 

15 domains and V L domains, V H domains only and V L domains only. 

Synergy in a therapeutic context may be achieved in a number of ways. For example, 
target combinations may be therapeutically active only if both targets are targeted by the 
ligand, whereas targeting one target alone is not therapeutically effective. In another 
20 embodiment, one target alone may provide some low or minimal therapeutic effect, but 
together with a second target the combination provides a synergistic increase in 
therapeutic effect. 

Preferably, the cytokines bound by the dual specific ligands of this aspect of the invention 
25 are sleeted from the list shown in Annex 2. 

Moreover, dual specific ligands may be used in oncology applications, where one 
specificity targets CD89, which is expressed by cytotoxic cells, and the other is tumour 
specific. Examples of tumour antigens which may be targetted are given in Annex 3. 

30 

In one embodiment of the second configuration of the invention, the variable domains are 
derived from an antibody directed against the first and/or second antigen or epitope. In a 
preferred embodiment the variable domains are derived from a repertoire of single 



WO 2004/003019 PCT/GB2003/002804 

25 

variable antibody domains. In one example, the repertoire is a repertoire that is not 
created in an animal or a synthetic repertoire, hi another example, the single variable 
domains are not isolated (at least in part) by animal immunisation. Thus, the single 
domains can be isolated from a naive library. 

5 

The second configuration of the invention, in another aspect, provides a multi-specific 
ligand comprising a first epitope binding domain having a first epitope binding specificity 
and a non-complementary second epitope binding domain having a second epitope 
binding specificity. The first and second binding specificities may be the same or 
10 different. 



In a further aspect, the present invention provides a closed conformation multi-specific 
ligand comprising a first epitope binding domain having a first epitope binding specificity 
and a non-complementary second epitope binding domain having a second epitope 
15 binding specificity wherein the first and second binding specificities are capable of 
competing for epitope binding such that the closed conformation multi-specific ligand 
cannot bind both epitopes simultaneously. 



In a still further aspect, the invention provides open conformation ligands comprising 
20 non-complementary binding domains, wherein the deomains are specific for a different 
epitope on the same target. Such ligands bind to targets with increased avidity. 
Similarly, the invention provides multivalent ligands comprising non-complementary 
binding domains specific for the same epitope and directed to targets which comprise 
multiple copies of said epitope, such as IL-5, PDGF-AA, PDGF-BB, TGF beta, TGF 
25 beta2, TGF beta3 and TNFa, for eample human TNF Receptor 1 and human TNFoc. 



In a similar aspect, ligands according to the invention can be configured to bind 
individual epitopes with low affinity, such that binding to individual epitopes is not 
therapeutically significant; but the increased avidity resulting from binding to two 
30 epitopes provides a theapeutic benefit. In a perticular example, epitopes may be targetted 
which are present individually on normal cell types, but present together only on 
abnormal or diseased cells, such as tumour cells. In such a situaton, only the abnormal or 



WO 2004/003019 PCT/GB2003/002804 

26 

diseased cells are effectively targetted by the bispecific ligands according to the 
invention. 

Ligand specific for multiple copies of the same epitope, or adjacent epitopes, on the same 
5 target (known as chelating dAbs) may also be trimeric or polymeric (tertrameric or more) 
ligands comprising three, four or more non-complementary binding domains. For 
example, ligands may be constructed comprising three or four V H domains or V L 
domains. 

10 Moreover, ligands are provided which bind to multisubunit targets, wherein each binding 
domain is specific for a subunit of said target. The ligand may be dimeric, trimeric or 
polymeric. 

Preferably, the multi-specific ligands according to the above aspects of the invention are 
15 obtainable by the method of the first aspect of the invention. 

According to the above aspect of the second configuration of the invention, 
advantageously the first epitope binding domain and the second epitope binding domains 
are non-complementary immunoglobulin variable domains, as herein defined. That is 
20 either V h ~Vh or V L "V L variable domains. 

Chelating dAbs in particular may be prepared according to a preferred aspect of the 
invention, namely the use of anchor dAbs, in which a library of dimeric, trimeric or 
multimeric dAbs is constructed using a vector which comprises a constant dAb upstream 
25 or downstream of a linker sequence, with a repertoire of second, third and further dAbs 
being inserted on the other side of the linker. For example, the anchor or guiding dAb 
maybe TAR1-5 (Vk), TAR1-27(Vk), TAR2h-5(VH) or TAR2h-6(Vic). 

In alternative methodologies, the use of linkers may be avoided, for example by the use of 
30 non-covalent bonding or naturall affinity between binding domains such as V H and V K . 
The invention accordingly provides a method for preparing a chelating multimeric ligand 
comprising the steps of: 



WO 2004/003019 PCT/GB2003/002804 

27 

(a) providing a vector comprising a nucleic acid sequence encoding a single 
binding domain specific for a first epitope on a target; 

(b) providing a vector encoding a repertoire comprising second binding domains 
specific for a second epitope on said target, which epitope can be the same or different to 

5 the first epitope, said second epitope being adjacent to said first epitope; and 

(c) expressing said first and second binding domains; and 

(d) isolating those combinations of first and second binding domains which 
combine together to produce a target-binding dimer. 

10 The first and second epitopes are adjacent such that a multimeric ligand is capable of 
binding to both epitopes simultaneously. This provides the ligand with the advantages of 
increased avidity if binding. Where the epitopes are the same, the increased avidity is 
obtained by the presence of multiple copies of the epitope on the target, allowing at least 
two copies to be simultaneously bound in order to obtain the increased avidity effect. 

15 

The binding domains may be associated by several methods, as well as the use of linkers. 
For example, the binding domains may comprise cys residues, avidin and streptavidin 
groups or other means for non-covalent attachment post-synthesis; those combinations 
which bind to the target efficiently will be isolated. Alternatively, a linker may be present 
20 between the first and second binding domains, which are expressed as a single 
polypeptide from a single vector, which comprises the first binding domain, the linker 
and a repertoire of second binding domains, for instance as described above. 

In a preferred aspect, the first and second binding domains associate naturally when 
25 bound to antigen; for example, V H and V K domains, when bound to adjacent epitopes, will 
naturally associate in a three-way interaction to form a stable dimer. Such associated 
proteins can be isolated in a target binding assay. An advantage of this procedure is that 
only binding domains which bind to closely adjacent epitopes, in the correct 
conformation, will associate and thus be isolated as a result of their increased avidity for 
30 the target. 

In an alternative embodiment of the above aspect of the second configuration of the 
invention, at least one epitope binding domain comprises a non-immunoglobulin 'protein 



WO 2004/003019 PCT/GB2003/002804 

28 

scaffold 5 or c protein skeleton 5 as herein defined. Suitable non-immuno globulin protein 
scaffolds include but are not limited to any of those selected from the group consisting of: 
SpA, fibronectin, GroEL and other chaperones, lipocallin, CCTLA4 and affibodies, as set 
forth above. 

5 

According to the above aspect of the second configuration of the invention, 
advantageously, the epitope binding domains are attached to a 'protein skeleton 5 . 
Advantageously, a protein skeleton according to the invention is an immunoglobulin 
skeleton. 

10 

According to the present invention, the term 'immunoglobulin skeleton 5 refers to a 
protein which comprises at least one immunoglobulin fold and which acts as a nucleus for 
one or more epitope binding domains, as defined herein. 

15 Preferred immunoglobulin skeletons as herein defined includes any one or more of those 
selected from the following: an immunoglobulin molecule comprising at least (i) the CL 
(kappa or lambda subclass) domain of an antibody; or (ii) the CHI domain of an antibody 
heavy chain; an immunoglobulin molecule comprising the CHI and CH2 domains of an 
antibody heavy chain; an immunoglobulin molecule comprising the CHI, CH2 and CH3 

20 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL 
(kappa or lambda subclass) domain of an antibody. A hinge region domain may also be 
included. Such combinations of domains may, for example, mimic natural antibodies, 
such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab 5 ) 2 molecules. 
Those skilled in the art will be aware that this list is not intended to be exhaustive. 

25 

Linking of the skeleton to the epitope binding domains, as herein defined may be 
achieved at the polypeptide level, that is after expression of the nucleic acid encoding the 
skeleton and/or the epitope binding domains. Alternatively, the linking step may be 
performed at the nucleic acid level. Methods of linking a protein skeleton according to the 
30 present invention, to the one or more epitope binding domains include the use of protein 
chemistry and/or molecular biology techniques which will be familiar to those skilled in 
the art and are described herein. 



WO 2004/003019 PCT/GB2003/002804 

29 

Advantageously, the closed conformation multispecific ligand may comprise a first 
domain capable of binding a target molecule, and a second domain capable of binding a 
molecule or group which extends the half-life of the ligand. For example, the molecule or 
group may be a bulky agent, such as HSA or a cell matrix protein. As used herein, the 

5 phrase "molecule or group which extends the half-life of a ligand" refers to a molecule or 
chemical group which, when bound by a dual-specific ligand as described herein 
increases the in vivo half-life of such dual specific ligand when administered to an 
animal, relative to a ligand that does not bind that molecule or group. Examples of 
molecules or groups that extend the half-life of a ligand are described hereinbelow. In a 

10 preferred embodiment, the closed conformation multispecific ligand may be capable of 
binding the target molecule only on displacement of the half-life enhancing molecule or 
group. Thus, for example, a closed conformation multispecific ligand is maintained in 
circulation in the bloodstream of a subject by a bulky molecule such as HSA. When a 
target molecule is encountered, competition between the binding domains of the closed 

15 conformation multispecific ligand results in displacement of the HSA and binding of the 
target. 

Ligands according to any aspect of the present invention, as well as dAb monomers 
useful in constructing such ligands, may advantageously dissociate from their cognate 
target(s) with a Kd of 300nM to 5pM (ie, 3 x 10~ 7 to 5 x 10" 12 M), preferably 50nM 
to20pM, or 5nM to 200pM or InM to lOOpM, 1 x 10" 7 M or less, 1 x 10" 8 M or less, 1 x 
10" 9 M or less, 1 x 10" 10 M or less, 1 x 10" 11 M or less; and/or a K off rate constant of 5 x 
10" 1 to 1 x 10~ 7 S'\ preferably 1 x 10' 2 to 1 x 1(T 6 S" 1 , or 5 x 1(T 3 to 1 x 10' 5 S~\ or 5 x 10" 1 
S' 1 or less, or 1 x 10" 2 S" 1 or less, or 1 x 10" 3 S" 1 or less, or 1 x 10" 4 S" 1 or less, or 1 x 10~ 5 S' 1 
or less, or 1 x 10" 6 S~ l or less as determined by surface plasmon resonance. The Kd rate 
constand is defined as Koff/Kon- 

In particular the invention provides an anti-TNFoc dAb monomer (or dual specific ligand 
comprising such a dAb), homodimer, heterodimer or homotrimer ligand, wherein each 
30 dAb binds TNFa. The ligand binds to TNFce with a Kd of 300nM to 5pM (ie, 3 x 1 0' 7 to 
5 x 10" 12 M), preferably 50nM to 20pM, more preferably 5nM to 200pM and most 
preferably InM to lOOpM; expressed in an alternative manner, the Kd is 1 x 10' M or 
less, preferably 1 x 10" 8 M or less, more preferably 1 x 10* 9 M or less, advantageously 1 x 



20 



25 



WO 2004/003019 PCT/GB2003/002804 

30 

10~ 10 M or less and most preferably 1 x 10' 11 M or less; and/or a Koff rate constant of 5 x 
10" 1 to 1 x 10" 7 S" 1 , preferably 1 x 10~ 2 to 1 x 1CT 6 S'\ more preferably 5 x 1CT 3 to 1 x 1CT 5 
S" 1 , for example 5 x 10' 1 S' 1 or less, preferably 1 x 1(T 2 S" 1 or less, more preferably 1x10" 
3 S~ l or less, advantageously 1 x 10' 4 S" 1 or less, further advantageously 1 x 10' 5 S" 1 or less, 
5 and most preferably 1 x 10" 6 S" 1 or less, as detemiined by surface plasmon resonance. 

Preferably, the ligand neutralises TNFa in a standard L929 assay with an ND50 of 
500nM to 50pM, preferably or lOOnM to 50pM, advantageously lOnM to lOOpM, more 
preferably InM to lOOpM; for example 50nM or less, preferably 5nM or less, 
10 advantageously 500pM or less, more preferably 200pM or less and most preferably 
lOOpM or less. 

Preferably, the ligand inhibits binding of TNF alpha to TNF alpha Receptor I (p55 
receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably 
15 lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 
5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most 
preferably lOOpM or less. Preferably, the TNFa is Human TNFa. 

Furthermore, the invention provides a an anti-TNF Receptor I dAb monomer, or dual 
20 specific ligand comprising such a dAb, that binds to TNF Receptor I with alQ of 300nM 
to 5pM (ie, 3 x 10" 7 to 5 x 10~ 12 M), preferably 50nM to20pM, more preferably 5nM to 
200pM and most preferably InM to lOOpM, for example 1 x 10" 7 M or less, preferably 1 
x 10' 8 M or less, more preferably 1 x 10" 9 M or less, advantageously 1 x 10" 10 M or less 

1 7 

and most preferably 1 x 10" 11 M or less; and/or a Koff rate constant of 5 x 10" to 1 x 10" 
25 S' 1 , preferably 1 x 1(T 2 to 1 x 10* 6 S* 1 , more preferably 5 x 1CT 3 to 1 x 10~ 5 S" 1 , for 
example 5 x 10" 1 S" 1 or less, preferably 1 x 10" 2 S" 1 or less, advantageously 1 x 10" 3 S" 1 or 
less, more preferably 1 x 10" 4 S" 1 or less, still more preferably 1 x 10~ 5 S" 1 or less, and most 
preferably 1 x 10" 6 S" 1 or less as determined by surface plasmon resonance. 

30 Preferably, the dAb monomeror ligand neutralises TNFa in a standard assay (eg, the 
L929 or HeLa assays described herein) with an ND50 of 500nM to 50pM, preferably 
lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for 



WO 2004/003019 PCT/GB2003/002804 

31 

example 50nM or less, preferably 5nM or less, more preferably 500pM or less, 
advantageously 200pM or less, and most preferably lOOpM or less. 

Preferably, the dAb monomer or ligand inhibits binding of TNF alpha to TNF alpha 
5 Receptor I (p55 receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, 
more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or 
less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or 
less, and most preferably lOOpM or less. Preferably, the TNF Receptor I target is Human 
TNFa. 

10 

Furthermore, the invention provides a dAb monomer(or dual specific ligand comprising 
such a dAb) that binds to serum albumin (SA) with a Kd of InM to 500^M (ie, x 10" 9 to 
5 x 10" 4 ), preferably lOOnM to 10/iM. Preferably, for a dual specific ligand comprising a 
first anti-SA dAb and a second dAb to another target, the affinity (eg Kd and/or Ko ff as 

15 measured by surface plasmon resonance, eg using BiaCore) of the second dAb for its 
target is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 
100000, or 10000 to 100000 times) the affinity of the first dAb for SA. For example, the 
first dAb binds SA with an affinity of approximately IOjuM, while the second dAb binds 
its target with an affinity of lOOpM. Preferably, the serum albumin is human serum 

20 albumin (HSA). 

In one embodiment, the first dAb (or a dAb monomer) binds SA (eg, HSA) with aKjof 
approximately 50, preferably 70, and more preferably 100, 150 or 200 nM. 

25 The invention moreover provides dimers, trimers and polymers of the aforementioned 
dAb monomers, in accordance with the foregoing aspect of the present invention. 

Ligands according to the invention, including dAb monomers, dimers and trimers, can be 
linked to an antibody Fc region, comprising one or both of C H 2 and C H 3 domains, and 
30 optionally a hinge region. For example, vectors encoding ligands linked as a single 
nucleotide sequence to an Fc region may be used to prepare such polypeptides. 



WO 2004/003019 PCT/GB2003/002804 

32 

In a further aspect of the second configuration of the invention, the present invention 
provides one or more nucleic acid molecules encoding at least a multispecific ligand as 
herein defined. In one embodiment, the ligand is a closed conformation ligand. In 
another embodiment, it is an open conformation ligand. The multispecific ligand may be 
5 encoded on a single nucleic acid molecule; alternatively, each epitope binding domain 
may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a 
single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, or 
may be separately expressed and subsequently linked together, for example using 
chemical linking agents. Ligands expressed from separate nucleic acids will be linked 
1 0 together by appropriate means. 

The nucleic acid may further encode a signal sequence for export of the polypeptides 
from a host cell upon expression and may be fused with a surface component of a 
filamentous bacteriophage particle (or other component of a selection display system) 
15 upon expression. Leader sequences, which may be used in bacterial expresion and/or 
phage or phagemid display, include pelB, stll, ompA, phoA, bla and pelA. 

In a further aspect of the second configuration of the invention the present invention 
provides a vector comprising nucleic acid according to the present invention. 

20 

In a yet further aspect, the present invention provides a host cell transfected with a vector 
according to the present invention. 

Expression from such a vector may be configured to produce, for example on the surface 
25 of a bacteriophage particle, epitope binding domains for selection. This allows selection 
of displayed domains and thus selection of 'multispecific ligands' using the method of the 
present invention. 

In a preferred embodiment of the second configuration of the invention, the epitope 
30 binding domains are immunoglobulin variable regions and are selected from single 
domain V gene repertoires. Generally the repertoire of single antibody domains is 
displayed on the surface of filamentous bacteriophage. In a preferred embodiment each 
single antibody domain is selected by binding of a phage repertoire to antigen. 



WO 2004/003019 



33 



PCT/GB2003/002804 



The present invention further provides a kit comprising at least a multispecific ligand 
according to the present invention, which may be an open conformation or closed 
conformation ligand. Kits according to the invention may be, for example, diagnostic 
5 kits, therapeutic kits, kits for the detection of chemical or biological species, and the like. 

In a further aspect still of the second configuration of the invention, the present invention 
provides a homogenous immunoassay using a ligand according to the present invention. 

10 In a further aspect still of the second configuration of the invention, the present invention 
provides a composition comprising a closed conformation multispecific ligand, obtainable 
by a method of the present invention, and a pharmaceutically acceptable carrier, diluent or 
excipient. 

15 Moreover, the present invention provides a method for the treatment of disease using a 
'closed conformation multispecific ligand' or a composition according to the present 
invention. 

In a preferred embodiment of the invention the disease is cancer or an inflammatory 
20 disease, eg rheumatoid arthritis, asthma or Crohn's disease. 

In a further aspect of the second configuration of the invention, the present invention 
provides a method for the diagnosis, including diagnosis of disease using a closed 
conformation multispecific ligand, or a composition according to the present invention. 
25 Thus in general the binding of an analyte to a closed conformation multispecific ligand 
may be exploited to displace an agent, which leads to the generation of a signal on 
displacement. For example, binding of analyte (second antigen) could displace an 
enzyme (first antigen) bound to the antibody providing the basis for an immunoassay, 
especially if the enzyme were held to the antibody through its active site. 

30 

Thus in a final aspect of the second configuration, the present invention provides a 
method for detecting the presence of a target molecule, comprising: 



WO 2004/003019 PCT/GB2003/002804 

34 

(a) providing a closed conformation multispecific ligand bound to an agent, said ligand 
being specific for the target molecule and the agent, wherein the agent which is bound by 
the ligand leads to the generation of a detectable signal on displacement from the ligand; 
(b) exposing the closed conformation multispecific ligand to the target molecule; and 
5 (c) detecting the signal generated as a result of the displacement of the agent. 

According to the above aspect of the second configuration of the invention, 
advantageously, the agent is an enzyme, which is inactive when bound by the closed 
conformation multi-specific ligand. Alternatively, the agent may be any one or more 
10 selected from the group consisting of the following: the substrate for an enzyme, and a 
fluorescent, luminescent or chromogenic molecule which is inactive or quenched when 
bound by the ligand. 

Sequences similar or homologous (e.g., at least about 70% sequence identity) to the 
15 sequences disclosed herein are also part of the invention. In some embodiments, the 
sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 
94%, 95%, 96%, 97%, 98%, 99% or higher. At the nucleic acid level, the sequence 
identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 
97% 98%, 99% or higher. Alternatively, substantial identity exists when the nucleic acid 
20 segments will hybridize under selective hybridization conditions (e.g., very high 
stringency hybridization conditions), to the complement of the strand. The nucleic acids 
may be present in whole cells, in a cell lysate, or in a partially purified or substantially 
pure form. 

25 Calculations of "homology" or "sequence identity" or "similarity" between two 
sequences (the terms are used interchangeably herein) are performed as follows. The 
sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in 
one or both of a first and a second amino acid or nucleic acid sequence for optimal 
alignment and non-homologous sequences can be disregarded for comparison purposes). 

30 In a preferred embodiment, the length of a reference sequence aligned for comparison 
purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more 
preferably at least 60%, and even more preferably at least 70%, 80%, 90%, 100% of the 
length of the reference sequence. The amino acid residues or nucleotides at 



WO 2004/003019 PCT/GB2003/002804 

35 

corresponding amino acid positions or nucleotide positions are then compared. When a 
position in the first sequence is occupied by the same amino acid residue or nucleotide as 
the corresponding position in the second sequence, then the molecules are identical at that 
position (as used herein amino acid or nucleic acid "homology" is equivalent to amino 
5 acid or nucleic acid "identity"). The percent identity between the two sequences is a 
function of the number of identical positions shared by the sequences, taking into account 
the number of gaps, and the length of each gap, which need to be introduced for optimal 
alignment of the two sequences. 

10 Advantageously, the BLAST algorithm (version 2.0) is employed for sequence alignment, 
with parameters set to default values. The BLAST algorithm is described in detail at the 
world wide web site ("www") of the National Center for Biotechnology Information 
("ncbi") of the National Institutes of Health ("nih") of the U.S. government (".gov"), in 
the "/Blast/" directory, in the "blastjielp.html" file. The search parameters are defined as 

15 follows, and are advantageously set to the defined default parameters. 

BLAST (Basic Local Alignment Search Tool) is the heuristic search algorithm employed 
by the programs blastp, blastn, blastx, tblastn, and tblastx; these programs ascribe 
significance to their findings using the statistical methods of Karlin and Altschul, 1990, 
20 Proc. Natl. Acad. Sci. USA 87(6):2264-8 (see the "blast Jielp.html" file, as described 
above) with a few enhancements. The BLAST programs were tailored for sequence 
similarity searching, for example to identify homologues to a query sequence. The 
programs are not generally useful for motif-style searching. For a discussion of basic 
issues in similarity searching of sequence databases, see Altschul et al. (1994). 

25 

The five BLAST programs available at the National Center for Biotechnology 
Information web site perform the following tasks: 

"blastp" compares an amino acid query sequence against a protein sequence database; 
"blastn" compares a nucleotide query sequence against a nucleotide sequence database; 
30 "blastx" compares the six-frame conceptual translation products of a nucleotide query 
sequence (both strands) against a protein sequence database; 

"tblastn" compares a protein query sequence against a nucleotide sequence database 
dynamically translated in all six reading frames (both strands). 



WO 2004/003019 PCT/GB2003/002804 

36 

"tblastx" compares the six-frame translations of a nucleotide query sequence against the 
six-frame translations of a nucleotide sequence database. 
BLAST uses the following search parameters: 

HISTOGRAM Display a histogram of scores for each search; default is yes. (See 
5 parameter H in the BLAST Manual). 

DESCRIPTIONS Restricts the number of short descriptions of matching sequences 
reported to the number specified; default limit is 100 descriptions. (See parameter V in 
the manual page). See also EXPECT and CUTOFF. 

ALIGNMENTS Restricts database sequences to the number specified for which high- 
10 scoring segment pairs (HSPs) are reported; the default limit is 50. If more database 
sequences than this happen to satisfy the statistical significance threshold for reporting 
(see EXPECT and CUTOFF below), only the matches ascribed the greatest statistical 
significance are reported. (See parameter B in the BLAST Manual). 

EXPECT The statistical significance threshold for reporting matches against database 
15 sequences; the default value is 10, such that 10 matches are expected to be found merely 
by chance, according to the stochastic model of Karlin and Altschul (1990). If the 
statistical significance ascribed to a match is greater than the EXPECT threshold, the 
match will not be reported. Lower EXPECT thresholds are more stringent, leading to 
fewer chance matches being reported. Fractional values are acceptable. (See parameter E 

20 in the BLAST Manual). 

CUTOFF Cutoff score for reporting high-scoring segment pairs. The default value is 
calculated from the EXPECT value (see above). HSPs are reported for a database 
sequence only if the statistical significance ascribed to them is at least as high as would be 
ascribed to a lone HSP having a score equal to the CUTOFF value. Higher CUTOFF 

25 values are more stringent, leading to fewer chance matches being reported. (See 
parameter S in the BLAST Manual). Typically, significance thresholds can be more 
intuitively managed using EXPECT. 

MATRIX Specify an alternate scoring matrix for BLASTP, BLASTX, TBLASTN and 
TBLASTX. The default matrix is BLOSUM62 (Henikoff & Henikoff, 1992, Proc. Natl. 
30 Aacad. Sci. USA 89(22):10915-9). The valid alternative choices include: PAM40, 
PAM120, PAM250 and IDENTITY. No alternate scoring matrices are available for 
BLASTN; specifying the MATRIX directive in BLASTN requests returns an error 
response. 



WO 2004/003019 PCT/GB2003/002804 

37 

STRAND Restrict a TBLASTN search to just the top or bottom strand of the database 
sequences; or restrict a BLASTN, BLASTX or TBLASTX search to just reading frames 
on the top or bottom strand of the query sequence. 

FILTER Mask off segments of the query sequence that have low compositional 
5 complexity, as determined by the SEG program of Wootton & Federhen (1993) 
Computers and Chemistry 17:149-163, or segments consisting of short-periodicity 
internal repeats, as determined by the XNU program of Claverie & States, 1993, 
Computers and Chemistry 17:191-201, or, for BLASTN, by the DUST program of 
Tatusov and Lipman (see the world wide web site of the NCBI). Filtering can eliminate 
10 statistically significant but biologically uninteresting reports from the blast output (e.g., 
hits against common acidic-, basic- or proline-rich regions), leaving the more biologically 
interesting regions of the query sequence available for specific matching against database 
sequences. 

15 Low complexity sequence found by a filter program is substituted using the letter "N" in 
nucleotide sequence (e.g., "N" repeated 13 times) and the letter "X" in protein sequences 
(e.g., "X" repeated 9 times). 

Filtering is only applied to the query sequence (or its translation products), not to 
20 database sequences. Default filtering is DUST for BLASTN, SEG for other programs. 

It is not unusual for nothing at all to be masked by SEG, XNU, or both, when applied to 
sequences in SWISS-PROT, so filtering should not be expected to always yield an effect. 
Furthermore, in some cases, sequences are masked in their entirety, indicating that the 
statistical significance of any matches reported against the unfiltered query sequence 
25 should be suspect. 

NCBI-gi Causes NCBI gi identifiers to be shown in the output, in addition to the 
accession and/or locus name. 

30 Most preferably, sequence comparisons are conducted using the simple BLAST search 
algorithm provided at the NCBI world wide web site described above, in the "/BLAST" 
directory. 



WO 2004/003019 



38 



PCT/GB2003/002804 



Brief Description of the Figures 

Figure 1 shows the diversification of V H /HSA at positions H50, H52, H52a, H53, 

H55, H56, H58, H95, H96, H97, H98 (DVT or NNK encoded 
5 respectively) which are in the antigen binding site of Vh HSA. The 

sequence of Vk is diversified at positions L50, L53. 

Figure 2 shows Library 1 : Germline Vk/DVT Vh, 

Library 2: Germline Vk/NNK V h , 
10 Library 3: Germline V H /DVT Vk 

Library 4: Germline V H /NNK V K 
In phage display/ScFv format. These libraries were pre-selected for 
binding to generic ligands protein A and protein L so that the majority of 
the clones and selected libraries are functional. Libraries were selected on 
15 HSA (first round) and jS-gal (second round) or HSA /3-gal selection or on 

/3-gal (first round) and HSA (second round) /3-gal HSA selection. Soluble 
scFv from these clones of PCR are amplified in the sequence. One clone 
encoding a dual specific antibody K8 was chosen for further work. 



20 Figure 3 shows an alignment of V H chains and V K chains. 

Figure 4 shows the characterisation of the binding properties of the K8 antibody, 

the binding properties of the K8 antibody characterised by monoclonal 
phage ELISA, the dual specific K8 antibody was found to bind HSA and 
25 /3-gal and displayed on the surface of the phage with absorbant signals 

greater than 1.0. No cross reactivity with other proteins was detected. 

Figure 5 shows soluble scFv ELISA performed using known concentrations of the 

K8 antibody fragment. A 96-well plate was coated with 100/xg of HSA, 
30 BSA and /3-gal at 10/ig/ml and 100/xg/ml of Protein A at 1/xg/ml 

concentration. 50/xg of the serial dilutions of the K8 scFv was applied and 
the bound antibody fragments were detected with Protein L-HRP. ELISA 
results confirm the dual specific nature of the K8 antibody. 



WO 2004/003019 



39 



PCT/GB2003/002804 



Figure 6 shows the binding characteristics of the clone K8V K /dummy V H analysed 

using soluble scFv ELISA. Production of the soluble scFv fragments was 
induced by IPTG as described by Harrison et al, Methods Enzymol. 
5 1996;267:83-109 and the supernatant containing scFv assayed directly. 

Soluble scFv ELISA is performed as described in example 1 and the bound 
scFvs were detected with Protein L-HRP. The ELISA results revealed that 
this clone was still able to bind /3-gal, whereas binding BSA was abolished. 

10 Figure 7 shows the sequence of variable domain vectors 1 and 2. 

Figure 8 is a map of the C H vector used to construct a V H 1/V H 2 multipsecific 

ligand. 

1 5 Figure 9 is a map of the V K vector used to construct a V K 1/V K 2 multispecific ligand. 

Figure 10 TNF receptor assay comparing TAJR1-5 dimer 4 5 TAR1-5-19 dimer 4 and 

TAR1-5-19 monomer. 

20 Figure 1 1 TNF receptor assay comparing TAR1-5 dimers 1-6. All dimers have been 

FPLC purified and the results for the optimal dimeric species are shown. 

Figure 12 TNF receptor assay of TAR1-5 19 homodimers in different formats: dAb- 

linker-dAb format with 3U, 5U or 7U linker, Fab format and cysteine 
25 hinge linker format. 

Figure 13 Dummy VH sequence for library 1. The sequence of the VH framework 

based on germline sequence DP47 - JH4b. Positions where NNK 
randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) 
30 has been incorporated into library 1 are indicated in bold underlined text. 



Figure 14 



Dummy VH sequence for library 2. The sequence of the VH framework 
based on germline sequence DP47 - JH4b. Positions where NNK 



WO 2004/003019 PCT/GB2003/002804 

40 

randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) 
has been incorporated into library 2 are indicated in bold underlined text. 



Figure 15 Dummy V/c sequence for library 3. The sequence of the Vk framework 

based on germline sequence DP K 9 - J K l. Positions where NNK 
randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) 
has been incorporated into library 3 are indicated in bold underlined text. 



Figure 16 Nucleotide and amino acid sequence of anti MSA dAbs MSA 16 and MSA 
10 26. 



Figure 17 Inhibition biacore of MSA 16 and 26. Purified dAbs MSA16 and MSA26 

were analysed by inhibition biacore to determine K d . Briefly, the dAbs 
were tested to determine the concentration of dAb required to achieve 

15 200RUs of response on a biacore CMS chip coated with a high density of 

MSA. Once the required concentrations of dAb had been determined, 
MSA antigen at a range of concentrations around the expected Kd was 
premixed with the dAb and incubated overnight. Binding to the MSA 
coated biacore chip of dAb in each of the premixes was then measured at a 

20 high flow-rate of 30 /xl/minute. 

Figure 1 8 Serum levels of MSA16 following injection. Serum half life of the dAb 

MSA16 was determined in mouse. MSA16 was dosed as single i.v. 
injections at approx 1.5mg/kg into GDI mice. Modelling with a 2 
25 compartment model showed MSA16 had a tl/2a of 0.98hr 5 a tl/2/3 of 

36.5hr and an AUG of 913hr.mg/ml. MSA16 had a considerably 
lengthened half life compared with HEL4 (an anti-hen egg white lysozyme 
dAb) which had a tl/2a of 0.06hr and a tl/2/3 of 0.34hr. 



30 Figure 19 ELISA (a) and TNF receptor assay (c) showing inhibition of TNF binding 

with a Fab-like fragment comprising MSA26Ck and TAR1-5-19CH. 
Addition of MSA with the Fab-like fragment reduces the level of 
inhibition. An ELISA plate coated with 1 jtig/ml TNFa was probed with 



WO 2004/003019 PCT/GB2003/002804 

41 

dual specific Vk C h and Vk Ck Fab like fragment and also with a control 
TNFa binding dAb at a concentration calculated to give a similar signal on 
the ELISA. Both the dual specific and control dAb were used to probe the 
ELISA plate in the presence and in the absence of 2mg/ml MSA. The 

5 signal in the dual specific well was reduced by more than 50% but the 

signal in the dAb well was not reduced at all (see figure 19a). The same 
dual specific protein was also put into the receptor assay with and without 
MSA and competition by MSA was also shown (see figure 19c). This 
demonstrates that binding of MSA to the dual specific is competitive with 

10 binding to TNF<x 



Figure 20 TNF receptor assay showing inhibiton of TNF binding with a disulphide 

bonded heterodimer of TAR1-5-19 dAb and MSA16 dAb. Addition of 
MSA with the dimer reduces the level of inhibiton in a dose dependant 
maimer. The TNF receptor assay (figure 19 (b)) was conducted in the 
presence of a constant concentration of heterodimer (18nM) and a dilution 
series of MSA and HSA. The presence of HSA at a range of 
concentrations (up to 2 mg/ml) did not cause a reduction in the ability of 
the dimer to inhibit TNFce . However, the addition of MSA caused a dose 
dependant reduction in the ability of the dimer to inhibit TNFa (figure 
19a) .This demonstrates that MSA and TNFa compete for binding to the 
cys bonded TAR1-5-19, MSA16 dimer. MSA and HSA alone did not 
have an effect on the TNF binding level in the assay. 



25 

Detailed Description of the Invention 
Definitions 

30 Complementary Two immunoglobulin domains are "complementary" where they 
belong to families of structures which form cognate pairs or groups or are derived from 
such families and retain this feature. For example, a V H domain and a V L domain of an 
antibody are complementary; two V H domains are not complementary, and two V L 



WO 2004/003019 PCT/GB2003/002804 

42 

domains are not complementary. Complementary domains may be found in other 
members of the immuno globulin superfamily, such as the V a and Vp (or y and S) domains 
of the T-cell receptor. In the context of the second configuration of the present invention, 
non-complementary domains do not bind a target molecule cooperatively, but act 
5 independently on different target epitopes which may be on the same or different 
molecules. Domains which are artificial, such as domains based on protein scaffolds 
which do not bind epitopes unless engineered to do so, are non-complementary. 
Likewise, two domains based on (for example) an immunoglobulin domain and a 
fibronectin domain are not complementary. 

10 

Immunoglobulin This refers to a family of polypeptides which retain the 
immunoglobulin fold characteristic of antibody molecules, which contains two (3 sheets 
and, usually, a conserved disulphide bond. Members of the immunoglobulin superfamily 
are involved in many aspects of cellular and non-cellular interactions in vivo, including 
15 widespread roles in the immune system (for example, antibodies, T-cell receptor 
molecules and the like), involvement in cell adhesion (for example the ICAM molecules) 
and intracellular signalling (for example, receptor molecules, such as the PDGF receptor). 
The present invention is applicable to all immunoglobulin superfamily molecules which 
possess binding domains. Preferably, the present invention relates to antibodies. 

20 

Combining Variable domains according to the invention are combined to form a group 
of domains; for example, complementary domains may be combined, such as V L domains 
being combined with V H domains. Non-complementary domains may also be combined. 
Domains may be combined in a number of ways, involving linkage of the domains by 
25 covalent or non-covalent means. 

Domain A domain is a folded protein structure which retains its tertiary structure 

independently of the rest of the protein. Generally, domains are responsible for discrete 
functional properties of proteins, and in many cases may be added, removed or 
30 transferred to other proteins without loss of function of the remainder of the protein 
and/or of the domain. By single antibody variable domain is meant a folded polypeptide 
domain comprising sequences characteristic of antibody variable domains. It therefore 
includes complete antibody variable domains and modified variable domains, for example 



WO 2004/003019 PCT/GB2003/002804 

43 

in which one or more loops have been replaced by sequences which are not characteristic 
of antibody variable domains, or antibody variable domains which have been truncated or 
comprise N- or C-terminal extensions, as well as folded fragments of variable domains 
which retain at least in part the binding activity and specificity of the full-length domain. 

5 

Repertoire A collection of diverse variants, for example polypeptide variants which 
differ in their primary sequence. A library used in the present invention will encompass a 
repertoire of polypeptides comprising at least 1000 members. 

10 Library The term library refers to a mixture of heterogeneous polypeptides or 

nucleic acids. The library is composed of members, each of which have a single 
polypeptide or nucleic acid sequence. To this extent, library is synonymous with 
repertoire. Sequence differences between library members are responsible for the 
diversity present in the library. The library may take the form of a simple mixture of 

15 polypeptides or nucleic acids, or may be in the form of organisms or cells, for example 
bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic 
acids. Preferably, each individual organism or cell contains only one or a limited number 
of library members. Advantageously, the nucleic acids are incorporated into expression 
vectors, in order to allow expression of the polypeptides encoded by the nucleic acids. In 

20 a preferred aspect, therefore, a library may take the form of a population of host 
organisms, each organism containing one or more copies of an expression vector 
containing a single member of the library in nucleic acid form which can be expressed to 
produce its corresponding polypeptide member. Thus, the population of host organisms 
has the potential to encode a large repertoire of genetically diverse polypeptide variants. 

25 

A 'closed conformation multi-specific ligand* describes a multi-specific ligand as 
herein defined comprising at least two epitope binding domains as herein defined. The 
term 'closed conformation' (multi-specific ligand) means that the epitope binding 
domains of the ligand are arranged such that epitope binding by one epitope binding 
30 domain competes with epitope binding by another epitope binding domain. That is, 
cognate epitopes may be bound by each epitope binding domain individually but not 
simultaneosuly. The closed conformation of the ligand can be achieved using methods 
herein described. 



WO 2004/003019 PCT/GB2003/002804 

44 

Antibody An antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such 
as a Fab , F(ab') 2? Fv, disulphide linked Fv, scFv, closed conformation multispecific 
antibody, disulphide-linked scFv, diabody) whether derived from any species naturally 
producing an antibody, or created by recombinant DNA technology; whether isolated 
5 from serum, B-cells, hybridomas, transfectomas, yeast or bacteria). 

Dual-specific ligand A ligand comprising a first immunoglobulin single variable domain 
and a second immunoglobulin single variable domain as herein defined, wherein the 
variable regions are capable of binding to two different antigens or two epitopes on the 

10 same antigen which are not normally bound by a monospecific immunoglobulin. For 
example, the two epitopes may be on the same hapten, but are not the same epitope or 
sufficiently adjacent to be bound by a monospecific ligand. The dual specific ligands 
according to the invention are composed of variable domains which have different 
specificities, and do not contain mutually complementary variable domain pairs which 

15 have the same specificity. 

Antigen A molecule that is bound by a ligand according to the present invention. 

Typically, antigens are bound by antibody ligands and are capable of raising an antibody 
response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule. 
20 Generally, the dual specific ligands according to the invention are selected for target 
specificity against a particular antigen. In the case of conventional antibodies and 
fragments thereof, the antibody binding site defined by the variable loops (LI, L2, L3 and 
HI, H2, H3) is capable of binding to the antigen. 

25 Epitope A unit of structure conventionally bound by an immunoglobulin V H /V L 
pair. Epitopes define the minimum binding site for an antibody, and thus represent the 
target of specificity of an antibody. In the case of a single domain antibody, an epitope 
represents the unit of structure bound by a variable domain in isolation. 

30 Generic ligand A ligand that binds to all members of a repertoire. Generally, not bound 
through the antigen binding site as defined above. Non-limiting examples include protein 
A, protein L and protein G. 



WO 2004/003019 PCT/GB2003/002804 

45 

Selecting Derived by screening, or derived by a Darwinian selection process, in 
which binding interactions are made between a domain and the antigen or epitope or 
between an antibody and an antigen or epitope. Thus a first variable domain may be 
selected for binding to an antigen or epitope in the presence or in the absence of a 
5 complementary variable domain. 

Universal framework A single antibody framework sequence corresponding to 

the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of 
Proteins of Immunological Interest", US Department of Health and Human Services) or 
10 corresponding to the human germline immunoglobulin repertoire or structure as defined 
by Chothia and Lesk, (1987) J. Mol. Biol. 196:910-917. The invention provides for the 
use of a single framework, or a set of such frameworks, which has been found to permit 
the derivation of virtually any binding specificity though variation in the hypervariable 
regions alone. 

15 

Half-life The time taken for the serum concentration of the ligand to reduce by 50%, 
in vivo, for example due to degradation of the ligand and/or clearance or sequestration of 
the ligand by natural mechanisms. The ligands of the invention are stabilised in vivo and 
their half-life increased by binding to molecules which resist degradation and/or clearance 

20 or sequestration. Typically, such molecules are naturally occurring proteins which 
themselves have a long half-life in vivo. The half-life of a ligand is increased if its 
functional activity persists, in vivo, for a longer period than a similar ligand which is not 
specific for the half-life increasing molecule. Thus, a ligand specific for HSA and a target 
molecule is compared with the same ligand wherein the specificity for HSA is not 

25 present, that it does not bind HSA but binds another molecule. For example, it may bind 
a second epitope on the target molecule. Typically, the half life is increased by 10%, 
20%o, 30%, 40%, 50% or more. Increases in the range of 2x, 3x, 4x, 5x, lOx, 20x, 30x, 
40x, 5 Ox or more of the half life are possible. Alternatively, or in addition, increases in 
the range of up to 30x, 40x, 50x, 60x, 70x, 80x, 90x, lOOx, 150x of the half life are 

30 possible. 

Homogeneous immunoassay An immunoassay in which analyte is detected 

without need for a step of separating bound and un-bound reagents. 



WO 2004/003019 



46 



PCT/GB2003/002804 



Substantially identical (or "substantially homologous") A first amino acid or 

nucleotide sequence that contains a sufficient number of identical or equivalent (e.g., with 
a similar side chain, e.g., conserved amino acid substitutions) amino acid residues or 
5 nucleotides to a second amino acid or nucleotide sequence such that the first and second 
amino acid or nucleotide sequences have similar activities. In the case of antibodies, the 
second antibody has the same binding specificity and has at least 50% of the affinity of 
the same. 

10 As used herein, the terms "low stringency " "medium stringency," "high stringency," 
or "very high stringency conditions" describe conditions for nucleic acid hybridization 
and washing. Guidance for performing hybridization reactions can be found in Current 
Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is 
incorporated herein by reference in its entirety. Aqueous and nonaqueous methods are 

15 described in that reference and either can be used. Specific hybridization conditions 
referred to herein are as follows: (1) low stringency hybridization conditions in 6X 
sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X 
SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C 
for low stringency conditions); (2) medium stringency hybridization conditions in 6X 

20 SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 60°C; (3) 
high stringency hybridization conditions in 6X SSC at about 45°C, followed by one or 
more washes in 0.2X SSC, 0.1% SDS at 65°C; and preferably (4) very high stringency 
hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one 
or more washes at 0.2X SSC, 1% SDS at 65°C. Very high stringency conditions (4) are 

25 the preferred conditions and the ones that should be used unless otherwise specified. 

Detailed Description of the Invention 

30 Unless defined otherwise, all technical and scientific terms used herein have the same 
meaning as commonly understood by one of ordinary skill in the art (e.g., in cell culture, 
molecular genetics, nucleic acid chemistry, hybridisation techniques and biochemistry). 
Standard techniques are used for molecular, genetic and biochemical methods (see 



WO 2004/003019 PCT/GB2003/002804 

47 

generally, Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d ed. (1989) 
Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. and Ausubel et al, Short 
Protocols in Molecular Biology (1999) 4 th Ed, John Wiley & Sons, Inc. which are 
incorporated herein by reference) and chemical methods. 

Preparation of immunoglobulin based multi-specific ligands 

Dual specific ligands according to the invention, whether open or closed in conformation 
according to the desired configuration of the invention, may be prepared according to 
previously established techniques, used in the field of antibody engineering, for the 
preparation of scFv, "phage" antibodies and other engineered antibody molecules. 
Techniques for the preparation of antibodies, and in particular bispecific antibodies, are 
for example described in the following reviews and the references cited therein: Winter & 
Milstein, (1991) Nature 349:293-299; Plueckthun (1992) Immunological Reviews 
130:151-188; Wright et al, (1992) Crti. Rev. Immunol.l2:125-168; Holliger, P. & 
Winter, G. (1993) Curr. Op. Biotechn. 4, 446-449; Carter, et al. (1995) J. Hematother. 4, 
463-470; Chester, K.A. & Hawkins, R.E. (1995) Trends Biotechn. 13, 294-300; 
Hoogenboom, H.R. (1997) Nature Biotechnol. 15, 125-126; Fearon, D. (1997) Nature 
Biotechnol. 15, 618-619; Pliickthun, A. & Pack, P. (1997) Immunotechnology 3, 83-105; 
Carter, P. & Merchant, A.M. (1997) Curr. Opin. Biotechnol. 8, 449-454; Holliger, P. & 
Winter, G. (1997) Cancer Immunol, hnmunother. 45,128-130. 

The invention provides for the selection of variable domains against two different 
antigens or epitopes, and subsequent combination of the variable domains. 

The techniques employed for selection of the variable domains employ libraries and 
selection procedures which are known in the art. Natural libraries (Marks et al (1991) J. 
Mol Biol, 222: 581; Vaughan et al (1996) Nature Biotech., 14: 309) which use 
rearranged V genes harvested from human B cells are well known to those skilled in the 
art. Synthetic libraries (Hoogenboom & Winter (1992) Mol. Biol, 227: 381; Barbas et 
al. (1992) Proc. Natl. Acad. Sci. USA, 89: 4457; Nissim et al (1994) EMBO J., 13: 692; 
Griffiths et al. (1994) EMBO J., 13: 3245; De Kruif et al. (1995) J. Mol. Biol, 248: 97) 
are prepared by cloning immunoglobulin V genes, usually using PCR. Errors in the PCR 



WO 2004/003019 PCT/GB2003/002804 

48 

process can lead to a high degree of randomisation. V H and/or V L libraries may be 
selected against target antigens or epitopes separately, in which case single domain 
binding is directly selected for, or together. 

5 A preferred method for making a dual specific ligand according to the present invention 
comprises using a selection system in which a repertoire of variable domains is selected 
for binding to a first antigen or epitope and a repertoire of variable domains is selected for 
binding to a second antigen or epitope. The selected variable first and second variable 
domains are then combined and the dual-specific ligand selected for binding to both first 
10 and second antigen or epitope. Closed conformation ligands are selected for binding both 
first and second antigen or epitope in isolation but not simultaneously. 

A. Library vector systems 

15 A variety of selection systems are known in the art which are suitable for use in the 
present invention. Examples of such systems are described below. 

Bacteriophage lambda expression systems may be screened directly as bacteriophage 
plaques or as colonies of lysogens, both as previously described (Huse et ah (1989) 
20 Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl. Acad. Set U.S.A., 87; 
Mullinax et ah (1990) Proc. Natl Acad. Sci. U.S.A., 87: 8095; Persson et al (1991) Proc. 
Natl Acad. Sci. U.S.A., 88: 2432) and are of use in the invention. Whilst such expression 
systems can be used to screen up to 10 6 different members of a library, they are not really 
suited to screening of larger numbers (greater than 10 6 members). 

25 

Of particular use in the construction of libraries are selection display systems, which 
enable a nucleic acid to be linked to the polypeptide it expresses. As used herein, a 
selection display system is a system that permits the selection, by suitable display means, 
of the individual members of the library by binding the generic and/or target ligands. 

30 

Selection protocols for isolating desired members of large libraries are known in the art, 
as typified by phage display techniques. Such systems, in which diverse peptide 
sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith 



WO 2004/003019 PCT/GB2003/002804 

49 

(1990) Science, 249: 386), have proven useful for creating libraries of antibody 
fragments (and the nucleotide sequences that encoding them) for the in vitro selection and 
amplification of specific antibody fragments that bind a target antigen (McCafferty et aL, 
WO 92/01047). The nucleotide sequences encoding the V H and V L regions are linked to 

5 gene fragments which encode leader signals that direct them to the periplasmic space of 
E. coli and as a result the resultant antibody fragments are displayed on the surface of the 
bacteriophage, typically as fusions to bacteriophage coat proteins (e.g., pill or pVIII). 
Alternatively, antibody fragments are displayed externally on lambda phage capsids 
(phagebodies). An advantage of phage-based display systems is that, because they are 
10 biological systems, selected library members can be amplified simply by growing the 
phage containing the selected library member in bacterial cells. Furthermore, since the 
nucleotide sequence that encode the polypeptide library member is contained on a phage 
or phagemid vector, sequencing, expression and subsequent genetic manipulation is 
relatively straightforward. 

15 

Methods for the construction of bacteriophage antibody display libraries and lambda 
phage expression libraries are well known in the art (McCafferty et aL (1990) Nature, 
348: 552; Kang et aL (1991) Proc. Natl Acad. Set U.S.A., 88: 4363; Clackson et aL 

(1991) Nature, 352: 624; Lowman et aL (1991) Biochemistry, 30: 10832; Burton et aL 
20 (1991) Proc. NatL Acad. Sci U.S.A., 88: 10134; Hoogenboom et aL (1991) Nucleic Acids 

Res., 19: 4133; Chang et aL (1991) J. Immunol., 147: 3610; Breitling et aL (1991) Gene, 
104: 147; Marks et al. (1991) supra; Barbas et aL (1992) supra; Hawkins and Winter 

(1992) J. Immunol., 22: 867; Marks et aL, 1992, J. Biol. Chem., 267: 16007; Lerner et aL 
(1992) Science, 258: 1313, incorporated herein by reference). 

25 

One particularly advantageous approach has been the use of scFv phage-libraries (Huston 
et aL, 1988, Proc. Natl. Acad. Sci U.S.A., 85: 5879-5883; Chaudhary et al. (1990) Proc. 
Natl. Acad. Sci U.S.A., 87: 1066-1070; McCafferty et aL (1990) supra; Clackson et aL 

(1991) Nature, 352: 624; Marks et aL (1991) J. Mol. Biol, 222: 581; Chiswell et aL 
30 (1992) Trends Biotech., 10: 80; Marks et al. (1992) J. Biol Chem., 267). Various 

embodiments of scFv libraries displayed on bacteriophage coat proteins have been 
described. Refinements of phage display approaches are also known, for example as 



WO 2004/003019 PCT/GB2003/002804 

50 

described in WO96/06213 and WO92/01047 (Medical Research Council et al) and 
WO97/08320 (Morphosys), which are incorporated herein by reference. 

Other systems for generating libraries of polypeptides involve the use of cell-free 
5 enzymatic machinery for the in vitro synthesis of the library members. In one method, 
RNA molecules are selected by alternate rounds of selection against a target ligand and 
PCR amplification (Tuerk and Gold (1990) Science, 249: 505; Ellington and Szostak 
(1990) Nature, 346: 818). A similar technique may be used to identify DNA sequences 
which bind a predetermined human transcription factor (Thiesen and Bach (1990) Nucleic 
10 Acids Res., 18: 3203; Beaudry and Joyce (1992) Science, 257: 635; WO92/05258 and 
W092/14843). In a similar way, in vitro translation can be used to synthesise 
polypeptides as a method for generating large libraries. These methods which generally 
comprise stabilised polysome complexes, are described further in WO88/08453, 
WO90/05785, WO90/07003, WO91/02076, WO91/05058, and WO92/02536. Alternative 
15 display systems which are not phage-based, such as those disclosed in W095/22625 and 
W095/1 1922 (Affymax) use the polysomes to display polypeptides for selection. 

A still further category of techniques involves the selection of repertoires in artificial 
compartments, which allow the linkage of a gene with its gene product. For example, a 

20 selection system in which nucleic acids encoding desirable gene products may be selected 
in microcapsules formed by water-in-oil emulsions is described in WO99/02671, 
WO00/40712 and Tawfik & Griffiths (1998) Nature Biotechnol 16(7), 652-6. Genetic 
elements encoding a gene product having a desired activity are compartmentalised into 
microcapsules and then transcribed and/or translated to produce their respective gene 

25 products (RNA or protein) within the microcapsules. Genetic elements which produce 
gene product having desired activity are subsequently sorted. This approach selects gene 
products of interest by detecting the desired activity by a variety of means. 

30 B. Library Construction . 

Libraries intended for selection, may be constructed using techniques known in the art, 
for example as set forth above, or may be purchased from commercial sources. Libraries 



WO 2004/003019 PCT/GB2003/002804 

51 

which are useful in the present invention are described, for example, in WO99/20749. 
Once a vector system is chosen and one or more nucleic acid sequences encoding 
polypeptides of interest are cloned into the library vector, one may generate diversity 
within the cloned molecules by undertaking mutagenesis prior to expression; 
alternatively, the encoded proteins may be expressed and selected, as described above, 
before mutagenesis and additional rounds of selection are performed. Mutagenesis of 
nucleic acid sequences encoding structurally optimised polypeptides is carried out by 
standard molecular methods. Of particular use is the polymerase chain reaction, or PCR, 
(Mullis and Faloona (1987) Methods Enzymol, 155: 335, herein incorporated by 
reference). PCR, which uses multiple cycles of DNA replication catalysed by a 
thermostable, DNA-dependent DNA polymerase to amplify the target sequence of 
interest, is well known in the art. The construction of various antibody libraries has been 
discussed in Winter et al. (1994) Ann. Rev. Immunology 12, 433-55, and references cited 
therein. 

PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at 
least 25 pmol of oligonucleotide primers; it may be advantageous to use a larger amount 
of primer when the primer pool is heavily heterogeneous, as each sequence is represented 
by only a small fraction of the molecules of the pool, and amounts become limiting in the 
later amplification cycles. A typical reaction mixture includes: 2ul of DNA, 25 pmol of 
oligonucleotide primer, 2.5 pi of 10X PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 
ul of 1.25 uM dNTP, 0.15 pi (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, 
Foster City, CA) and deionized water to a total volume of 25 pi. Mineral oil is overlaid 
and the PCR is performed using a programmable thermal cycler. The length and 
temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in 
accordance to the stringency requirements in effect. Annealing temperature and timing 
are determined both by the efficiency with which a primer is expected to anneal to a 
template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid 
molecules are simultaneously amplified and mutagenised, mismatch is required, at least 
in the first round of synthesis. The ability to optimise the stringency of primer annealing 
conditions is well within the knowledge of one of moderate skill in the art. An annealing 
temperature of between 30 °C and 72 °C is used. Initial denaturation of the template 
molecules normally occurs at between 92°C and 99°C for 4 minutes, followed by 20-40 



WO 2004/003019 PCT/GB2003/002804 

52 

cycles consisting of denaturation (94-99°C for 15 seconds to 1 minute), annealing 
(temperature determined as discussed above; 1-2 minutes), and extension (72°C for 1-5 
minutes, depending on the length of the amplified product). Final extension is generally 
for 4 minutes at 72°C, and may be followed by an indefinite (0-24 hour) step at 4°C. 

C. Combining single variable domains 

Domains useful in the invention, once selected, may be combined by a variety of methods 
known in the art, including covalent and non-covalent methods. 

Preferred methods include the use of polypeptide linkers, as described, for example, in 
connection with scFv molecules (Bird et al, (1988) Science 242:423-426). Discussion of 
suitable linkers is provided in Bird et al. Science 242, 423-426; Hudson et al , Journal 
Immunol Methods 231 (1999) 177-189; Hudson et al, Proc Nat Acad Sci USA 85, 5879- 
5883. Linkers are preferably flexible, allowing the two single domains to interact. One 
linker example is a (Gly 4 Ser) n linker, where n=l to 8, eg, 2, 3, 4, 5 or 7. The linkers used 
in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) PNAS 
(USA) 90:6444-6448). 

In one embodiment, the linker employed is not an immunoglobulin hinge region. 

Variable domains may be combined using methods other than linkers. For example, the 
use of disulphide bridges, provided through naturally-occurring or engineered cysteine 
residues, may be exploited to stabilise V h -Vh>V l -Vl or V H -V L dimers (Reiter et al, 
(1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable 
domains to improve the "fit" and thus the stability of interaction (Ridgeway et al, (1996) 
Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788). 

Other techniques for joining or stabilising variable domains of immunoglobulins, and in 
particular antibody V H domains, may be employed as appropriate. 

In accordance with the present invention, dual specific ligands can be in "closed" 
conformations in solution. A "closed" configuration is that in which the two domains (for 



WO 2004/003019 PCT/GB2003/002804 

53 

example V H and V L ) are present in associated form, such as that of an associated V H -V L 
pair which forms an antibody binding site. For example, scFv may be in a closed 
conformation, depending on the arrangement of the linker used to link the V H and V L 
domains. If this is sufficiently flexible to allow the domains to associate, or rigidly holds 
5 them in the associated position, it is likely that the domains will adopt a closed 
conformation. 

Similarly, V H domain pairs and V L domain pairs may exist in a closed conformation. 
Generally, this will be a function of close association of the domains, such as by a rigid 
10 linker, in the ligand molecule. Ligands in a closed conformation will be unable to bind 
both the molecule which increases the half-life of the ligand and a second target molecule. 
Thus, the ligand will typically only bind the second target molecule on dissociation from 
the molecule which increases the half-life of the ligand. 

15 Moreover, the construction of V H /V H , Vi/Vl or V H /V L dimers without linkers provides 
for competition between the domains. 

Ligands according to the invention may moreover be in an open conformation. In such a 
conformation, the ligands will be able to simultaneously bind both the molecule which 

20 increases the half-life of the ligand and the second target molecule. Typically, variable 
domains in an open configuration are (in the case of V H -V L pairs) held far enough apart 
for the domains not to interact and form an antibody binding site and not to compete for 
binding to their respective epitopes. In the case of V H /V H or Vi/V L dimers, the domains 
are not forced together by rigid linkers. Naturally, such domain pairings will not compete 

25 for antigen binding or form an antibody binding site. 

Fab fragments and whole antibodies will exist primarily in the closed conformation, 
although it will be appreciated that open and closed dual specific ligands are likely to 
exist in a variety of equilibria under different circumstances. Binding of the ligand to a 
30 target is likely to shift the balance of the equilibrium towards the open configuration. 
Thus, certain ligands according to the invention can exist in two conformations in 
solution, one of which (the open form) can bind two antigens or epitopes independently, 



WO 2004/003019 PCT/GB2003/002804 

54 

whilst the alternative conformation (the closed form) can only bind one antigen or 
epitope; antigens or epitopes thus compete for binding to the ligand in this conformation. 

Although the open form of the dual specific ligand may thus exist in equilibrium with the 
5 closed form in solution, it is envisaged that the equilibrium will favour the closed form; 
moreover, the open form can be sequestered by target binding into a closed conformation. 
Preferably, therefore, certain dual specific ligands of the invention are present in an 
equilibrium between two (open and closed) conformations. 

10 Dual specific ligands according to the invention may be modified in order to favour an 
open or closed conformation. For example, stabilisation of V H -V L interactions with 
disulphide bonds stabilises the closed conformation. Moreover, linkers used to join the 
domains, including V H domain and V L domain pairs, may be constructed such that the 
open from is favoured; for example, the linkers may sterically hinder the association of 

15 the domains, such as by incorporation of large amino acid residues in opportune 
locations, or the designing of a suitable rigid structure which will keep the domains 
physically spaced apart. 

D. Characterisation of the dual-specific ligand . 

20 

The binding of the dual-specific ligand to its specific antigens or epitopes can be tested by 
methods which will be familiar to those skilled in the art and include ELISA. In a 
preferred embodiment of the invention binding is tested using monoclonal phage ELISA. 

25 Phage ELISA may be performed according to any suitable procedure: an exemplary 
protocol is set forth below. 

Populations of phage produced at each round of selection can be screened for binding by 
ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. 
30 Phage from single infected bacterial colonies from these populations can then be screened 
by ELISA to identify "monoclonal" phage antibodies. It is also desirable to screen soluble 
antibody fragments for binding to antigen or epitope, and this can also be undertaken by 



WO 2004/003019 PCT/GB2003/002804 

55 

ELISA using reagents, for example, against a C- or N-terminal tag (see for example 
Winter et al (1994) Ann. Rev. Immunology 12, 433-55 and references cited therein. 

The diversity of the selected phage monoclonal antibodies may also be assessed by gel 
5 electrophoresis of PGR products (Marks et al 1991, supra; Nissim et al 1994 supra), 
probing (Tomlinson et al, 1992) J. Mol. Biol. 227, 776) or by sequencing of the vector 
DNA. 

E. Structure of 'Dual-specific ligands' . 

As described above, an antibody is herein defined as an antibody (for example IgG, IgM, 
IgA, IgA, IgE) or fragment (Fab, Fv, disulphide linked Fv, scFv, diabody) which 
comprises at least one heavy and a light chain variable domain, at least two heavy chain 
variable domains or at least two light chain variable domains. It may be at least partly 
derived from any species naturally producing an antibody, or created by recombinant 
DNA technology; whether isolated from serum, B -cells, hybridomas, transfectomas, yeast 
or bacteria). 

In a preferred embodiment of the invention the dual-specific ligand comprises at least one 
20 single heavy chain variable domain of an antibody and one single light chain variable 
domain of an antibody, or two single heavy or light chain variable domains. For example, 
the ligand may comprise a V H /V L pair, a pair of V H domains or a pair of V L domains. 

The first and the second variable domains of such a ligand may be on the same 
25 polypeptide chain. Alternatively they may be on separate polypeptide chains. In the case 
that they are on the same polypeptide chain they may be linked by a linker, which is 
preferentially a peptide sequence, as described above. 

The first and second variable domains may be covalently or non-covalently associated. In 
30 the case that they are covalently associated, the covalent bonds may be disulphide bonds. 

In the case that the variable domains are selected from V-gene repertoires selected for 
instance using phage display technology as herein described, then these variable domains 



10 



15 



WO 2004/003019 PCT/GB2003/002804 

56 

comprise a universal framework region, such that is they may be recognised by a specific 
generic ligand as herein defined. The use of universal frameworks, generic ligands and 
the like is described in WO99/20749. 

5 Where V-gene repertoires are used variation in polypeptide sequence is preferably located 
within the structural loops of the variable domains. The polypeptide sequences of either 
variable domain may be altered by DNA shuffling or by mutation in order to enhance the 
interaction of each variable domain with its complementary pair. DNA shuffling is 
known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and 
10 U.S. Patent No. 6,297,053, both of which are incorporated herein by reference. Other 
methods of mutagenesis are well known to those of skill in the art. 

In a preferred embodiment of the invention the 'dual-specific ligand 5 is a single chain Fv 
fragment. In an alternative embodiment of the invention, the ' dual-specific ligand' 
15 consists of a Fab format. 

In a further aspect, the present invention provides nucleic acid encoding at least a c dual- 
specific ligand' as herein defined. 

20 One skilled in the art will appreciate that, depending on the aspect of the invention, both 
antigens or epitopes may bind simultaneously to the same antibody molecule. 
Alternatively, they may compete for binding to the same antibody molecule. For 
example, where both epitopes are bound simultaneously, both variable domains of a dual 
specific ligand are able to independently bind their target epitopes. Where the domains 

25 compete, the one variable domain is capable of binding its target, but not at the same time 
as the other variable domain binds its cognate target; or the first variable domain is 
capable of binding its target, but not at the same time as the second variable domain binds 
its cognate target. 

30 The variable regions may be derived from antibodies directed against target antigens or 
epitopes. Alternatively they may be derived from a repertoire of single antibody domains 
such as those expressed on the surface of filamentous bacteriophage. Selection may be 
performed as described below. 



WO 2004/003019 



PCT/GB2003/002804 



57 



In general, the nucleic acid molecules and vector constructs required for the performance 
of the present invention may be constructed and manipulated as set forth in standard 
laboratory manuals, such as Sambrook et ah (1989) Molecular Cloning: A Laboratory 
5 Manual, Cold Spring Harbor, USA. 

The manipulation of nucleic acids useful in the present invention is typically carried out 
in recombinant vectors. 

10 Thus in a further aspect, the present invention provides a vector comprising nucleic acid 
encoding at least a 'dual-specific ligand' as herein defined. 

As used herein, vector refers to a discrete element that is used to introduce heterologous 
DNA into cells for the expression and/or replication thereof. Methods by which to select 

15 or construct and, subsequently, use such vectors are well known to one of ordinary skill in 
the art. Numerous vectors are publicly available, including bacterial plasmids, 
bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used 
for simple cloning and mutagenesis; alternatively gene expression vector is employed. A 
vector of use according to the invention may be selected to accommodate a polypeptide 

20 coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb or more in 
length A suitable host cell is transformed with the vector after in vitro cloning 
manipulations. Each vector contains various functional components, which generally 
include a cloning (or "polylinker") site, an origin of replication and at least one selectable 
marker gene. If given vector is an expression vector, it additionally possesses one or more 

25 of the following: enhancer element, promoter, transcription termination and signal 
sequences, each positioned in the vicinity of the cloning site, such that they are 
operatively linked to the gene encoding a ligand according to the invention. 

Both cloning and expression vectors generally contain nucleic acid sequences that enable 
30 the vector to replicate in one or more selected host cells. Typically in cloning vectors, this 
sequence is one that enables the vector to replicate independently of the host 
chromosomal DNA and includes origins of replication or autonomously replicating 
sequences. Such sequences are well known for a variety of bacteria, yeast and viruses. 



WO 2004/003019 PCT/GB2003/002804 

58 

The origin of replication from the plasmid pBR322 is suitable for most Gram-negative 
bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g. 
SV 40, adenovirus) are useful for cloning vectors in mammalian cells. Generally, the 
origin of replication is not needed for mammalian expression vectors unless these are 
5 used in mammalian cells able to replicate high levels of DNA, such as COS cells. 

Advantageously, a cloning or expression vector may contain a selection gene also 
referred to as selectable marker. This gene encodes a protein necessary for the survival or 
growth of transformed host cells grown in a selective culture medium. Host cells not 
10 transformed with the vector containing the selection gene will therefore not survive in the 
culture medium. Typical selection genes encode proteins that confer resistance to 
antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, 
complement auxotrophic deficiencies, or supply critical nutrients not available in the 
growth media. 

15 

Since the replication of vectors encoding a ligand according to the present invention is 
most conveniently performed in E. coli, an E. co/z-selectable marker, for example, the p- 
lactamase gene that confers resistance to the antibiotic ampicillin, is of use. These can be 
obtained from E. coli plasmids, such as pBR322 or a pUC plasmid such as pUC18 or 
20 pUC19. 

Expression vectors usually contain a promoter that is recognised by the host organism and 
is operably linked to the coding sequence of interest. Such a promoter may be inducible 
or constitutive. The term "operably linked" refers to a juxtaposition wherein the 
25 components described are in a relationship permitting them to function in their intended 
manner. A control sequence "operably linked" to a coding sequence is ligated in such a 
way that expression of the coding sequence is achieved under conditions compatible with 
the control sequences. 

30 Promoters suitable for use with prokaryotic hosts include, for example, the (3-lactamase 
and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system 
and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems 



WO 2004/003019 PCT/GB2003/002804 

59 

will also generally contain a Shine-Delgarno sequence operably linked to the coding 
sequence. 

The preferred vectors are expression vectors that enables the expression of a nucleotide 
5 sequence corresponding to a polypeptide library member. Thus, selection with the first 
and/or second antigen or epitope can be performed by separate propagation and 
expression of a single clone expressing the polypeptide library member or by use of any 
selection display system. As described above, the preferred selection display system is 
bacteriophage display. Thus, phage or phagemid vectors may be used, eg pITl or pIT2. 

10 Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pel A. 
One example are phagemid vectors which have an E. colt origin of replication (for 
double stranded replication) and also a phage origin of replication (for production of 
single-stranded DNA). The manipulation and expression of such vectors is well known in 
the art (Hoogenboom and Winter (1992) supra; Nissim et aL (1994) supra). Briefly, the 

15 vector contains a P -lactamase gene to confer selectivity on the phagemid and a lac 
promoter upstream of a expression cassette that consists (N to C terminal) of a pelB 
leader sequence (which directs the expressed polypeptide to the periplasmic space), a 
multiple cloning site (for cloning the nucleotide version of the library member), 
optionally, one or more peptide tag (for detection), optionally, one or more TAG stop 

20 codon and the phage protein plIL Thus, using various suppressor and non-suppressor 
strains of E. coli and with the addition of glucose, iso-propyl thio-(3~D-galactoside (EPTG) 
or a helper phage, such as VCS Ml 3, the vector is able to replicate as a plasmid with no 
expression, produce large quantities of the polypeptide library member only or produce 
phage, some of which contain at least one copy of the polypeptide-pIII fusion on their 

25 surface. 

Construction of vectors encoding ligands according to the invention employs 
conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, 
tailored, and religated in the form desired to generate the required vector. If desired, 
30 analysis to confirm that the correct sequences are present in the constructed vector can be 
performed in a known fashion. Suitable methods for constructing expression vectors, 
preparing in vitro transcripts, introducing DNA into host cells, and performing analyses 
for assessing expression and function are known to those skilled in the art. The presence 



WO 2004/003019 PCT/GB2003/002804 

60 

of a gene sequence in a sample is detected, or its amplification and/or expression 
quantified by conventional methods, such as Southern or Northern analysis, Western 
blotting, dot blotting of DNA, RNA or protein, in situ hybridisation, 
immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those 
5 skilled in the art will readily envisage how these methods may be modified, if desired. 

Structure of closed conformation multispecific lisands 

According to one aspect of the second configuration of the invention present invention, 
10 the two or more non- complementary epitope binding domains are linked so that they are 
in a closed conformation as herein defined. Advantageously, they may be further 
attached to a skeleton which may, as a alternative, or on addition to a linker described 
herein, facilitate the formation and/or maintenance of the closed conformation of the 
epitope binding sites with respect to one another. 

15 

(I) Skeletons 

Skeletons may be based on immunoglobulin molecules or may be non-immunoglobulin in 
origin as set forth above. Preferred immunoglobulin skeletons as herein defined includes 
any one or more of those selected from the following: an immunoglobulin molecule 

20 comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the 
CHI domain of an antibody heavy chain; an immunoglobulin molecule comprising the 
CHI and CH2 domains of an antibody heavy chain; an immunoglobulin molecule 
comprising the CHI, CH2 and CH3 domains of an antibody heavy chain; or any of the 
subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody. 

25 A hinge region domain may also be included.. Such combinations of domains may, for 
example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, 
scFv, Fab or F(ab') 2 molecules. Those skilled in the art will be aware that this list is not 
intended to be exhaustive. 

30 (II) Protein scaffolds 

Each epitope binding domain comprises a protein scaffold and one or more CDRs which 
are involved in the specific interaction of the domain with one or more epitopes. 
Advantageously, an epitope binding domain according to the present invention comprises 



WO 2004/003019 PCT/GB2003/002804 

61 

three CDRs. Suitable protein scaffolds include any of those selected from the group 
consisting of the following: those based on immunoglobulin domains, those based on 
fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones 
such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors 
5 SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be 
exhaustive. 

F: Scaffolds for use in Constructing Dual Specific Ligands 

10 i. Selection of the main-chain conformation 

The members of the immunoglobulin superfamily all share a similar fold for their 
polypeptide chain. For example, although antibodies are highly diverse in terms of their 
primary sequence, comparison of sequences and cryst alio graphic structures has revealed 
that, contrary to expectation, five of the six antigen binding loops of antibodies (HI, H2, 

15 LI, L2, L3) adopt a limited number of main-chain conformations, or canonical structures 
(Chothia and Lesk (1987) J. Mol Biol, 196: 901; Chothia et al (1989) Nature, 342: 877). 
Analysis of loop lengths and key residues has therefore enabled prediction of the main- 
chain conformations of HI, H2, LI, L2 and L3 found in the majority of human antibodies 
(Chothia et al (1992) J. Mol Biol, 227: 799; Tomlinson et ah (1995) EMBO J., 14: 

20 4628; Williams et al (1996) J. Moh Biol, 264: 220). Although the H3 region is much 
more diverse in terms of sequence, length and structure (due to the use of D segments), it 
also forms a limited number of main-chain conformations for short loop lengths which 
depend on the length and the presence of particular residues, or types of residue, at key 
positions in the loop and the antibody framework (Martin et al (1996) J. Mol Biol, 263: 

25 800; Shirai et al (1996) FEBS Letters, 399: 1). 

The dual specific ligands of the present invention are advantageously assembled from 
libraries of domains, such as libraries of Vh domains and/or libraries of Vl domains. 
Moreover, the dual specific ligands of the invention may themselves be provided in the 
30 form of libraries. In one aspect of the present invention, libraries of dual specific ligands 
and/or domains are designed in which certain loop lengths and key residues have been 
chosen to ensure that the main-chain conformation of the members is known. 
Advantageously, these are real conformations of immunoglobulin superfamily molecules 



WO 2004/003019 PCT/GB2003/002804 

62 

found in nature, to minimise the chances that they are non-functional, as discussed 
above. Germline V gene segments serve as one suitable basic framework for constructing 
antibody or T-cell receptor libraries; other sequences are also of use. Variations may 
occur at a low frequency, such that a small number of functional members may possess an 
5 altered main-chain conformation, which does not affect its function. 



Canonical structure theory is also of use to assess the number of different main-chain 
conformations encoded by ligands, to predict the main-chain conformation based on 
ligand sequences and to chose residues for diversification which do not affect the 

10 canonical structure. It is known that, in the human V K domain, the LI loop can adopt one 
of four canonical structures, the L2 loop has a single canonical structure and that 90% of 
human V K domains adopt one of four or five canonical structures for the L3 loop 
(Tomlinson et aL (1995) supra); thus, in the V K domain alone, different canonical 
structures can combine to create a range of different main-chain conformations. Given 

15 that the domain encodes a different range of canonical structures for the LI, L2 and L3 
loops and that V K and V\ domains can pair with any Vh domain which can encode several 
canonical structures for the HI and H2 loops, the number of canonical structure 
combinations observed for these five loops is very large. This implies that the generation 
of diversity in the main-chain conformation may be essential for the production of a wide 

20 range of binding specificities. However, by constructing an antibody library based on a 
single known main-chain conformation it has been found, contrary to expectation, that 
diversity in the main-chain conformation is not required to generate sufficient diversity to 
target substantially all antigens. Even more surprisingly, the single main-chain 
conformation need not be a consensus structure - a single naturally occurring 

25 conformation can be used as the basis for an entire library. Thus, in a preferred aspect, the 
dual-specific ligands of the invention possess a single known main-chain conformation. 



The single main-chain conformation that is chosen is preferably commonplace among 
molecules of the immunoglobulin superfamily type in question. A conformation is 
30 commonplace when a significant number of naturally occurring molecules are observed 
to adopt it. Accordingly, in a preferred aspect of the invention, the natural occurrence of 
the different main-chain conformations for each binding loop of an immunoglobulin 
domain are considered separately and then a naturally occurring variable domain is 



WO 2004/003019 PCT/GB2003/002804 

63 

chosen which possesses the desired combination of main-chain conformations for the 
different loops. If none is available, the nearest equivalent may be chosen. It is preferable 
that the desired combination of main-chain conformations for the different loops is 
created by selecting germline gene segments which encode the desired main-chain 
5 conformations. It is more preferable, that the selected germline gene segments are 
frequently expressed in nature, and most preferable that they are the most frequently 
expressed of all natural germline gene segments. 



In designing dual specific ligands or libraries thereof the incidence of the different main- 

10 chain conformations for each of the six antigen binding loops may be considered 
separately. For HI, H2, LI, L2 and L3, a given conformation that is adopted by between 
20% and 100% of the antigen binding loops of naturally occurring molecules is chosen. 
Typically, its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, 
above 50% or even above 65%. Since the vast majority of H3 loops do not have canonical 

15 structures, it is preferable to select a main-chain conformation which is commonplace 
among those loops which do display canonical structures. For each of the loops, the 
conformation which is observed most often in the natural repertoire is therefore selected. 
In human antibodies, the most popular canonical structures (CS) for each loop are as 
follows: HI - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), LI - CS 2 of 

20 V K (39%), L2 - CS 1 (100%), L3 - CS 1 of V K (36%) (calculation assumes a k:A, ratio of 
70:30, Hood et al. (1967) Cold Spring Harbor Symp. Quant BioL, 48: 133). For H3 loops 
that have canonical structures, a CDR3 length (Kabat et al. (1991) Sequences of proteins 
of immunological interest, U.S. Department of Health and Human Services) of seven 
residues with a salt-bridge from residue 94 to residue 101 appears to be the most 

25 common. There are at least 16 human antibody sequences in the EMBL data library with 
the required H3 length and key residues to form this conformation and at least two 
crystallographic structures in the protein data bank which can be used as a basis for 
antibody modelling (2cgr and ltet). The most frequently expressed germline gene 
segments that this combination of canonical structures are the Vh segment 3-23 (DP-47), 

30 the J H segment JH4b, the V K segment 02/012 (DPK9) and the J K segment J K 1. V H 
segments DP45 and DP38 are also suitable. These segments can therefore be used in 
combination as a basis to construct a library with the desired single main-chain 
conformation. 



WO 2004/003019 



64 



PCT/GB2003/002804 



Alternatively, instead of choosing the single main-chain conformation based on the 
natural occurrence of the different main-chain conformations for each of the binding 
loops in isolation, the natural occurrence of combinations of main-chain conformations is 
used as the basis for choosing the single main-chain conformation. In the case of 
antibodies, for example, the natural occurrence of canonical structure combinations for 
any two, three, four, five or for all six of the antigen binding loops can be determined. 
Here, it is preferable that the chosen conformation is commonplace in naturally occurring 
antibodies and most preferable that it observed most frequently in the natural repertoire. 
Thus, in human antibodies, for example, when natural combinations of the five antigen 
binding loops, HI, H2, LI, L2 and L3, are considered, the most frequent combination of 
canonical structures is determined and then combined with the most popular 
conformation for the H3 loop, as a basis for choosing the single main-chain conformation. 

ii. Diversification of the canonical sequence 

Having selected several known main-chain conformations or, preferably a single 
known main-chain conformation, dual specific ligands according to the invention or 
libraries for use in the invention can be constructed by varying the binding site of the 
molecule in order to generate a repertoire with structural and/or functional diversity. This 
means that variants are generated such that they possess sufficient diversity in their 
structure and/or in their function so that they are capable of providing a range of 
activities. 

The desired diversity is typically generated by varying the selected molecule at one or 
more positions. The positions to be changed can be chosen at random or are preferably 
selected. The variation can then be achieved either by randomisation, during which the 
resident amino acid is replaced by any amino acid or analogue thereof, natural or 
synthetic, producing a very large number of variants or by replacing the resident amino 
acid with one or more of a defined subset of amino acids, producing a more limited 
number of variants. 

Various methods have been reported for introducing such diversity. Error-prone PCR 
(Hawkins et al (1992) J. Mol Biol., 226: 889), chemical mutagenesis (Deng et ah (1994) 



WO 2004/003019 PCT/GB2003/002804 

65 

J. Biol Chem., 269: 9533) or bacterial mutator strains (Low et al (1996) X Mol Biol, 
260: 359) can be used to introduce random mutations into the genes that encode the 
molecule. Methods for mutating selected positions are also well known in the art and 
include the use of mismatched oligonucleotides or degenerate oligonucleotides, with or 
5 without the use of PGR. For example, several synthetic antibody libraries have been 
created by targeting mutations to the antigen binding loops. The H3 region of a human 
tetanus toxoid-binding Fab has been randomised to create a range of new binding 
specificities (Barbas et al (1992) Proc. Natl Acad. Set USA, 89: 4457). Random or 
semi-random H3 and L3 regions have been appended to germline V gene segments to 

10 produce large libraries with unmutated framework regions (Hoogenboom & Winter 
(1992) J. Mol Biol, 227: 381; Barbas et al (1992) Proc. Natl Acad. Set USA, 89: 4457; 
Nissim et al (1994) EMBO J., 13: 692; Griffiths et al (1994) EMBO J., 13: 3245; De 
Kraif et al (1995) J. Mol. Biol, 248: 97). Such diversification has been extended to 
include some or all of the other antigen binding loops (Crameri et al (1996) Nature Med., 

15 2: 100; Riechmann et al (1995) Bio/Technology, 13: 475; Morphosys, WO97/08320, 
supra). 

Since loop randomisation has the potential to create approximately more than 10 15 
structures for H3 alone and a similarly large number of variants for the other five loops, it 
20 is not feasible using current transformation technology or even by using cell free systems 
to produce a library representing all possible combinations. For example, in one of the 

in 

largest libraries constructed to date, 6 x 10 different antibodies, which is only a fraction 
of the potential diversity for a library of this design, were generated (Griffiths et al. 
(1994) supra). 

25 

In a preferred embodiment, only those residues which are directly involved in creating or 
modifying the desired function of the molecule are diversified. For many molecules, the 
function will be to bind a target and therefore diversity should be concentrated in the 
target binding site, while avoiding changing residues which are crucial to the overall 
30 packing of the molecule or to maintaining the chosen main-chain conformation. 



WO 2004/003019 PCT/GB2003/002804 

66 

Diversification of the canonical sequence as it applies to antibody domains 

In the case of antibody dual-specific ligands, the binding site for the target is most 
often the antigen binding site. Thus, in a highly preferred aspect, the invention provides 
libraries of or for the assembly of antibody dual-specific ligands in which only those 
5 residues in the antigen binding site are varied. These residues are extremely diverse in the 
human antibody repertoire and are known to make contacts in high-resolution 
antibody/antigen complexes. For example, in L2 it is known that positions 50 and 53 are 
diverse in naturally occurring antibodies and are observed to make contact with the 
antigen. In contrast, the conventional approach would have been to diversify all the 
10 residues in the corresponding Complementarity Determining Region (CDR1) as defined 
by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the 
library for use according to the invention. This represents a significant improvement in 
terms of the functional diversity required to create a range of antigen binding specificities. 

15 In nature, antibody diversity is the result of two processes: somatic recombination of 
germline V, D and J gene segments to create a naive primary repertoire (so called 
germline and junctional diversity) and somatic hypermutation of the resulting rearranged 
V genes. Analysis of human antibody sequences has shown that diversity in the primary 
repertoire is focused at the centre of the antigen binding site whereas somatic 

20 hypermutation spreads diversity to regions at the periphery of the antigen binding site that 
are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. Mol. Biol, 
256: 813). This complementarity has probably evolved as an efficient strategy for 
searching sequence space and, although apparently unique to antibodies, it can easily be 
applied to other polypeptide repertoires. The residues which are varied are a subset of 

25 those that form the binding site for the target. Different (including overlapping) subsets of 
residues in the target binding site are diversified at different stages during selection, if 
desired. 

In the case of an antibody repertoire, an initial 'naive' repertoire is created where some, 
30 but not all, of the residues in the antigen binding site are diversified. As used herein in 
this context, the term "naive" refers to antibody molecules that have no pre-determined 
target. These molecules resemble those which are encoded by the immunoglobulin genes 
of an individual who has not undergone immune diversification, as is the case with fetal 



WO 2004/003019 PCT/GB2003/002804 

67 

and newborn individuals, whose immune systems have not yet been challenged by a 
wide variety of antigenic stimuli. This repertoire is then selected against a range of 
antigens or epitopes. If required, further diversity can then be introduced outside the 
region diversified in the initial repertoire. This matured repertoire can be selected for 
5 modified function, specificity or affinity. 



The invention provides two different naive repertoires of binding domains for the 
construction of dual specific ligands, or a naive library of dual specific ligands, in which 
some or all of the residues in the antigen binding site are varied. The "primary" library 

10 mimics the natural primary repertoire, with diversity restricted to residues at the centre of 
the antigen binding site that are diverse in the germline V gene segments (germline 
diversity) or diversified during the recombination process (junctional diversity). Those 
residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, 
H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96. In the "somatic" 

15 library, diversity is restricted to residues that are diversified during the recombination 
process (junctional diversity) or are highly somatically mutated). Those residues which 
are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, 
L31, L32, L34 and L96. All the residues listed above as suitable for diversification in 
these libraries are known to make contacts in one or more antibody-antigen complexes. 

20 Since in both libraries, not all of the residues in the antigen binding site are varied, 
additional diversity is incorporated during selection by varying the remaining residues, if 
it is desired to do so. It shall be apparent to one skilled in the art that any subset of any of 
these residues (or additional residues which comprise the antigen binding site) can be 
used for the initial and/or subsequent diversification of the antigen binding site. 

25 

In the construction of libraries for use in the invention, diversification of chosen positions 
is typically achieved at the nucleic acid level, by altering the coding sequence which 
specifies the sequence of the polypeptide such that a number of possible amino acids (all 
20 or a subset thereof) can be incorporated at that position. Using the IUPAC 
30 nomenclature, the most versatile codon is NNK, which encodes all amino acids as well as 
the TAG stop codon. The NNK codon is preferably used in order to introduce the 
required diversity. Other codons which achieve the same ends are also of use, including 



WO 2004/003019 PCT/GB2003/002804 

68 

the NNN codon, which leads to the production of the additional stop codons TGA and 
TAA. 

A feature of side-chain diversity in the antigen binding site of human antibodies is a 
5 pronounced bias which favours certain amino acid residues. If the amino acid 
composition of the ten most diverse positions in each of the V H , V K and V x regions are 
summed, more than 76% of the side-chain diversity comes from only seven different 
residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), 
alanine (7%), aspartate (6%) and threonine (6%). This bias towards hydrophilic residues 
10 and small residues which can provide main-chain flexibility probably reflects the 
evolution of surfaces which are predisposed to binding a wide range of antigens or 
epitopes and may help to explain the required promiscuity of antibodies in the primary 
repertoire. 



15 Since it is preferable to mimic this distribution of amino acids, the distribution of amino 
acids at the positions to be varied preferably mimics that seen in the antigen binding site 
of antibodies. Such bias in the substitution of amino acids that permits selection of certain 
polypeptides (not just antibody polypeptides) against a range of target antigens is easily 
applied to any polypeptide repertoire. There are various methods for biasing the amino 

20 acid distribution at the position to be varied (including the use of tri-nucleotide 
mutagenesis, see WO97/08320), of which the preferred method, due to ease of synthesis, 
is the use of conventional degenerate codons. By comparing the amino acid profile 
encoded by all combinations of degenerate codons (with single, double, triple and 
quadruple degeneracy in equal ratios at each position) with the natural amino acid use it is 

25 possible to calculate the most representative codon. The codons (AGT)(AGC)T, 
(AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using 
RJPAC nomenclature - are those closest to the desired amino acid profile: they encode 
22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and 
cysteine. Preferably, therefore, libraries are constructed using either the DVT, DVC or 

30 DVY codon at each of the diversified positions. 



WO 2004/003019 PCT/GB2003/002804 

69 

G: Antigens capable of increasing ligand half-life 

The dual specific ligands according to the invention, in one configuration thereof, are 
capable of binding to one or more molecules which can increase the half-life of the ligand 
5 in vivo. Typically, such molecules are polypeptides which occur naturally in vivo and 
which resist degradation or removal by endogenous mechanisms which remove unwanted 
material from the organism. For example, the molecule which increases the half-life of 
the organism may be selected from the following: 

10 Proteins from the extracellular matrix; for example collagen, laminins, integrins and 

fibronectin. Collagens are the major proteins of the extracellular matrix. About 15 types 
of collagen molecules are currently known, found in different parts of the body, eg type I 
collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, 
cornea, internal organs or type II collagen found in cartilage, invertebral disc, notochord, 

15 vitreous humour of the eye. 

Proteins found in blood, including: 

Plasma proteins such as fibrin, a~2 macro globulin, serum albumin, fibrinogen A, 
20 fibrinogen B, serum amyloid protein A, heptaglobin, profilin, ubiquitin, uteroglobulin and 
/^-microglobulin; 

Enzymes and inhibitors such as plasminogen, lysozyme, cystatin C, alpha- 1 -antitrypsin 
and pancreatic trypsin inhibitor. Plasminogen is the inactive precursor of the trypsin-like 
25 serine protease plasmin. It is normally found circulating through the blood stream. When 
plasminogen becomes activated and is converted to plasmin, it unfolds a potent enzymatic 
domain that dissolves the fibrinogen fibers that entgangle the blood cells in a blood clot. 
This is called fibrinolysis. 

30 Immune system proteins, such as IgE, IgG, IgM. 



Transport proteins such as retinol binding protein, a-l microglobulin. 



WO 2004/003019 PCT/GB2003/002804 

70 

Defensins such as beta-defensin 1, Neutrophil defensins 1,2 and 3. 

Proteins found at the blood brain barrier or in neural tissues, such as melanocortin 
receptor, myelin, ascorbate transporter. 

5 

Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see 
US5977307); 

brain capillary endothelial cell receptor, transferrin, transferrin receptor, insulin, insulin- 
10 like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, 
insulin receptor. 

Proteins localised to the kidney, such as polycystin, type IV collagen, organic anion 
transporter Kl, Heymann's antigen. 

15 

Proteins localised to the liver, for example alcohol dehydrogenase, G250. 

Blood coagulation factor X 
cd antitrypsin 
20 HNF la 

Proteins localised to the lung, such as secretory component (binds IgA). 

Proteins localised to the Heart, for example HSP 27. This is associated with dilated 
25 cardiomyopathy. 

Proteins localised to the skin, for example keratin. 

Bone specific proteins, such as bone morphogenic proteins (BMPs), which are a subset of 
30 the transforming growth factor |8 superfamily that demonstrate osteogenic activity. 

Examples include BMP-2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-1) and 
-8 (OP-2). 



WO 2004/003019 PCT/GB2003/002804 

71 

Tumour specific proteins, including human trophoblast antigen, herceptin receptor, 
oestrogen receptor, cathepsins eg cathepsin B (found in liver and spleen). 



Disease-specific proteins, such as antigens expressed only on activated T-cells: including 
5 LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL) see Nature 402, 
304-309; 1999, OX40 (a member of the TNF receptor family, expressed on activated T 
cells and the only costimulatory T cell molecule known to be specifically up-regulated in 
human T cell leukaemia virus type-I (HTLV-I) -producing cells.) See J Immunol 2000 
Jul 1;1 65(1): 263-70; Metalloproteases (associated with arthritis/cancers), including 
10 CG65 12 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; 
angiogenic growth factors, including acidic fibroblast growth factor (FGF-1), basic 
fibroblast growth factor (FGF-2), Vascular endothelial growth factor / vascular 
permeability factor (VEGF/VPF), transforming growth factor-a (TGF a), tumor necrosis 
factor-alpha (TNF-ce), angiogenic interleukin-3 (IL-3), interleukin-8 (IL-8), platelet- 
15 derived endothelial growth factor (PD-ECGF), placental growth factor (P1GF), midkine 
platelet-derived growth factor-BB (PDGF), fractalkine. 

Stress proteins (heat shock proteins) 

HSPs are normally found intracellularly. When they are found extracellularly, it is an 
20 indicator that a cell has died and spilled out its contents. This unprogrammed cell death 
(necrosis) only occurs when as a result of trauma, disease or injury and therefore in vivo, 
extracellular HSPs trigger a response from the immune system that will fight infection 
and disease. A dual specific which binds to extracellular HSP can be localised to a 
disease site. 

25 

Proteins involved in Fc transport 
Brambell receptor (also known as FcRB) 

This Fc receptor has two functions, both of which are potentially useful for delivery 
The functions are 

30 (1) The transport of IgG from mother to child across the placenta 

(2) the protection of IgG from degradation thereby prolonging its serum half life of 
IgG. It is thought that the receptor recycles IgG from endosome. 



WO 2004/003019 PCT/GB2003/002804 

72 

See Holliger et ah Nat Biotechnol 1997 Jul;15(7):632-6. 

Ligands according to the invention may designed to be specific for the above targets 
without requiring any increase in or increasing half life in vivo. For example, ligands 
according to the invention can be specific for targets selected from the foregoing which 
are tissue-specific, thereby enabling tissue-specific targeting of the dual specific ligand, 
or a dAb monomer that binds a tissue-specific therapeutically relevant target, irrespective 
of any increase in half-life, although this may result. Moreover, where the ligand or dAb 
monomer targets kidney or liver, this may redirect the ligand or dAb monomer to an 
alternative clearance pathway in vivo (for example, the ligand may be directed away from 
liver clearance to kidney clearance). 

H: Use of multispecific ligands according to the second configuration of the 
invention 

Multispecific ligands according to the method of the second configuration of the present 
invention may be employed in in vivo therapeutic and prophylactic applications, in vitro 
and in vivo diagnostic applications, in vitro assay and reagent applications, and the like. 
For example antibody molecules may be used in antibody based assay techniques, such as 
ELIS A techniques, according to methods known to those skilled in the art. 

As alluded to above, the multispecific ligands according to the invention are of use in 
diagnostic, prophylactic and therapeutic procedures. Multispecific antibodies according to 
the invention are of use diagnostically in Western analysis and in situ protein detection by 
standard immunohistochemical procedures; for use in these applications, the ligands may 
be labelled in accordance with techniques known to the art. In addition, such antibody 
polypeptides may be used preparatively in affinity chromatography procedures, when 
complexed to a chromatographic support, such as a resin. All such techniques are well 
known to one of skill in the art. 

Diagnostic uses of the closed conformation multispecific ligands according to the 
invention include homogenous assays for analytes which exploit the ability of closed 
conformation multispecific ligands to bind two targets in competition, such that two 



WO 2004/003019 PCT/GB2003/002804 

73 

targets cannot bind simultaneously (a closed conformation), or alternatively their ability 
to bind two targets simultaneously (an open conformation). 

A true homogenous immunoassay format has been avidly sought by manufacturers of 
5 diagnostics and research assay systems used in drug discovery and development. The 
main diagnostics markets include human testing in hospitals, doctor's offices and clinics, 
commercial reference laboratories, blood banks, and the home, non-human diagnostics 
(for example food testing, water testing, environmental testing, bio-defence, and 
veterinary testing), and finally research (including drug development; basic research and 
1 0 academic research) . 

At present all these markets utilise immunoassay systems that are built around 
chemiluminescent, ELISA, fluorescence or in rare cases radioimmunoassay 
technologies. Each of these assay formats requires a separation step (separating bound 

15 from un-bound reagents). In some cases, several separation steps are required. Adding 
these additional steps adds reagents and automation, takes time, and affects the ultimate 
outcome of the assays. In human diagnostics, the separation step may be automated, 
which masks the problem, but does not remove it. The robotics, additional reagents, 
additional incubation times, and the like add considerable cost and complexity. In drug 

20 development, such as high throughput screening, where literally millions of samples are 
tested at once, with very low levels of test molecule, adding additional separation steps 
can eliminate the ability to perform a screen. However, avoiding the separation creates 
too much noise in the read out. Thus, there is a need for a true homogenous format that 
provides sensitivities at the range obtainable from present assay formats. Advantageously, 

25 an assay possesses fully quantitative read-outs with high sensitivity and a large dynamic 
range. Sensitivity is an important requirement, as is reducing the amount of sample 
required. Both of these features are features that a homogenous system offers. This is 
very important in point of care testing, and in drug development where samples are 
precious. Heterogenous systems, as currently available in the art, require large quantities 

30 of sample and expensive reagents 

Applications for homogenous assays include cancer testing, where the biggest assay is 
that for Prostate Specific Antigen, used in screening men for prostate cancer. Other 



WO 2004/003019 PCT/GB2003/002804 

74 

applications include fertility testing, which provides a series of tests for women 
attempting to conceive including beta-hcg for pregnancy. Tests for infectious diseases, 
including hepatitis, HIV, rubella, and other viruses and microorganisms and sexually 
transmitted diseases. Tests are used by blood banks, especially tests for HIV, hepatitis A, 
5 B, C, non A non B. Therapeutic drug monitoring tests include monitoring levels of 
prescribed drugs in patients for efficacy and to avoid toxicity, for example digoxin for 
arrhythmia, and phenobarbital levels in psychotic cases; theophylline for asthma. 
Diagnostic tests are moreover useful in abused drug testing, such as testing for cocaine, 
marijuana and the like. Metabolic tests are used for measuring thyroid function, anaemia 
10 and other physiological disorders and functions. 

The homogenous immunoassay format is moreover useful in the manufacture of standard 
clinical chemistry assays. The inclusion of immunoassays and chemistry assays on the 
same instrument is highly advantageous in diagnostic testing. Suitable chemical assays 
15 include tests for glucose, cholesterol, potassium, and the like. 

A further major application for homogenous immunoassays is drug discovery and 
development: high throughput screening includes testing combinatorial chemistry 
libraries versus targets in ultra high volume. Signal is detected, and positive groups then 

20 split into smaller groups, and eventually tested in cells and then animals. Homogenous 
assays may be used in all these types of test. In drug development, especially animal 
studies and clinical trials heavy use of immunoassays is made. Homogenous assays 
greatly accelerate and simplify these procedures. Other Applications include food and 
beverage testing: testing meat and other foods for E. coli, salmonella, etc; water testing, 

25 including testing at water plants for all types of contaminants including E. coli; and 
veterinary testing. 

In a broad embodiment, the invention provides a binding assay comprising a detectable 
agent which is bound to a closed conformation multispecific ligand according to the 
30 invention, and whose detectable properties are altered by the binding of an analyte to said 
closed conformation multispecific ligand. Such an assay may be configured in several 
different ways, each exploiting the above properties of closed conformation multispecific 
ligands. 



WO 2004/003019 



PCT/GB2003/002804 



75 



The assay relies on the direct or indirect displacement of an agent by the analyte, resulting 
in a change in the detectable properties of the agent. For example, where the agent is an 
enzyme which is capable of catalysing a reaction which has a detectable end-point, said 
5 enzyme can be bound by the ligand such as to obstruct its active site, thereby inactivating 
the enzyme. The analyte, which is also bound by the closed conformation multispecific 
ligand, displaces the enzyme, rendering it active through freeing of the active site. The 
enzyme is then able to react with a substrate, to give rise to a detectable event. In an 
alternative embodiment, the ligand may bind the enzyme outside of the active site, 
10 influencing the conformation of the enzyme and thus altering its activity. For example, 
the structure of the active site may be constrained by the binding of the ligand, or the 
binding of cofactors necessary for activity may be prevented. 

The physical implementation of the assay may take any form known in the art. For 
15 example, the closed conformation multispecific ligand/enzyme complex may be provided 
on a test strip; the substrate may be provided in a different region of the test strip, and a 
solvent containing the analyte allowed to migrate through the ligand/enzyme complex, 
displacing the enzyme, and carrying it to the substrate region to produce a signal. 
Alternatively, the ligand/enzyme complex may be provided on a test stick or other solid 
20 phase, and dipped into an analyte/substrate solution, releasing enzyme into the solution in 
response to the presence of analyte. 

Since each molecule of analyte potentially releases one enzyme molecule, the assay is 
quantitative, with the strength of the signal generated in a given time being dependent on 
25 the concentration of analyte in the solution. 

Further configurations using the analyte in a closed conformation are possible. For 
example, the closed conformation multispecific ligand may be configured to bind an 
enzyme in an allosteric site, thereby activating the enzyme. In such an embodiment, the 
30 enzyme is active in the absence of analyte. Addition of the analyte displaces the enzyme 
and removes allosteric activation, thus inactivating the enzyme. 



WO 2004/003019 PCT/GB2003/002804 

76 

In the context of the above embodiments which employ enzyme activity as a measure of 
the analyte concentration, activation or inactivation of the enzyme refers to an increase or 
decrease in the activity of the enzyme, measured as the ability of the enzyme to catalyse a 
signal-generating reaction. For example, the enzyme may catalyse the conversion of an 
undetectable substrate to a detectable form thereof. For example, horseradish peroxidase 
is widely used in the art together with chromogenic or chemiluminescent substrates, 
which are available commercially. The level of increase or decrease of the activity of the 
enzyme may between 10% and 100%, such as 20%, 30%, 40%, 50%, 60%, 70%, 80% or 
90%; in the case of an increase in activity, the increase may be more than 100%, i.e. 
200%, 300%, 500% or more, or may not be measurable as a percentage if the baseline 
activity of the inhibited enzyme is undetectable. 

In a further configuration, the closed conformation multispecific ligand may bind the 
substrate of an enzyme/substrate pair, rather than the enzyme. The substrate is therefore 
unavailable to the enzyme until released from the closed conformation multispecific 
ligand through binding of the analyte. The implementations for this configuration are as 
for the configurations which bind enzyme. 

Moreover, the assay may be configured to bind a fluorescent molecule, such as a 
fluorescein or another fluorophore, in a conformation such that the fluorescence is 
quenched on binding to the ligand. In this case, binding of the analyte to the ligand will 
displace the fluorescent molecule, thus producing a signal. Alternatives to fluorescent 
molecules which are useful in the present invention include luminescent agents, such as 
luciferin/luciferase, and chromogenic agents, including agents commonly used in 
immunoassays such as BDRP. 

Therapeutic and prophylactic uses of multispecific ligands prepared according to the 
invention involve the administration of ligands according to the invention to a recipient 
mammal, such as a human. Multi-specificity can allow antibodies to bind to multimeric 
antigen with great avidity. Multispecific ligands can allow thecross- linking of two 
antigens, for example in recruiting cytotoxic T-cells to mediate the killing of tumour cell 
lines. 



WO 2004/003019 PCT/GB2003/002804 

77 

Substantially pure ligands or binding proteins thereof, for example dAb monomers, of at 
least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 
99% or more homogeneity is most preferred for pharmaceutical uses, especially when the 
mammal is a human. Once purified, partially or to homogeneity as desired, the ligands 
5 may be used diagnostically or therapeutically (including extracorporeally) or in 
developing and performing assay procedures, immunofluorescent stainings and the like 
(Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, 
Academic Press, NY). 

10 The ligands or binding proteins thereof, for example dAb monomers, of the present 
invention will typically find use in preventing, suppressing or treating inflammatory 
states, allergic hypersensitivity, cancer, bacterial or viral infection, and autoimmune 
disorders (which include, but are not limited to, Type I diabetes, asthma, multiple 
sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and 

15 myasthenia gravis). 

In the instant application, the term "prevention" involves administration of the protective 
composition prior to the induction of the disease. "Suppression" refers to administration 
of the composition after an inductive event, but prior to the clinical appearance of the 
20 disease. "Treatment" involves administration of the protective composition after disease 
symptoms become manifest. 

Animal model systems which can be used to screen the effectiveness of the antibodies or 
binding proteins thereof in protecting against or treating the disease are available. 

25 Methods for the testing of systemic lupus erythematosus (SLE) in susceptible mice are 
known in the art (Knight et al. (1978) J. Exp. Med., 147: 1653; Reinersten et al. (1978) 
New Eng. J. Med., 299: 515). Myasthenia Gravis (MG) is tested in SJL/J female mice by 
inducing the disease with soluble AchR protein from another species (Lindstrom et al. 
(1988) Adv. Immunol., 42: 233). Arthritis is induced in a susceptible strain of mice by 

30 injection of Type II collagen (Stuart et al. (1984) Ann. Rev. Immunol., 42: 233). A model 
by which adjuvant arthritis is induced in susceptible rats by injection of mycobacterial 
heat shock protein has been described (Van Eden et al. (1988) Nature, 331: 171). 
Thyroiditis is induced in mice by administration of thyroglobulin as described (Maron et 



WO 2004/003019 PCT/GB2003/002804 

78 

al (1980) J. Exp. Med., 152: 1115). Insulin dependent diabetes mellitus (IDDM) occurs 
naturally or can be induced in certain strains of mice such as those described by 
Kanasawa et al (1984) Diabetologia, 27: 113. EAE in mouse and rat serves as a model 
for MS in human. In this model, the demyelinating disease is induced by administration 
5 of myelin basic protein (see Paterson (1986) Textbook of Immunopathology, Mischer et 
al, eds., Grune and Stratton, New York, pp. 179-213; McFarlin et al (1973) Science, 
179: 478: and Satoh et al (1987) J. Immunol, 138: 179). 

Generally, the present ligands will be utilised in purified form together with 
10 pharmacologically appropriate carriers. Typically, these carriers include aqueous or 
alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or 
buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, 
dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable 
adjuvants, if necessary to keep a polypeptide complex in suspension, may be chosen from 
15 thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates. 

Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, 
such as those based on Ringer's dextrose. Preservatives and other additives, such as 
antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack 
20 (1 982) Remington 's Pharmaceutical Sciences, 1 6th Edition). 

The ligands of the present invention may be used as separately administered compositions 
or in conjunction with other agents. These can include various immunotherapeutic drugs, 
such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. 
25 Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents 
in conjunction with the ligands of the present invention, or even combinations of lignds 
according to the present invention having different specificities, such as ligands selected 
using different target antigens or epitopes, whether or not they are pooled prior to 
administration. 

30 

The route of administration of pharmaceutical compositions according to the invention 
may be any of those commonly known to those of ordinary skill in the art. For therapy, 
including without limitation immunotherapy, the selected ligands thereof of the invention 



WO 2004/003019 PCT/GB2003/002804 

79 

can be administered to any patient in accordance with standard techniques. The 
administration can be by any appropriate mode, including parenterally, intravenously, 
intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, 
appropriately, by direct infusion with a catheter. The dosage and frequency of 
5 administration will depend on the age, sex and condition of the patient, concurrent 
administration of other drugs, counterindications and other parameters to be taken into 
account by the clinician. 



The ligands of this invention can be lyophilised for storage and reconstituted in a suitable 
10 carrier prior to use. This technique has been shown to be effective with conventional 
immunoglobulins and art-known lyophilisation and reconstitution techniques can be 
employed. It will be appreciated by those skilled in the art that lyophilisation and 
reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional 
immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) 
15 and that use levels may have to be adjusted upward to compensate. 

The compositions containing the present ligands or a cocktail thereof can be administered 
for prophylactic and/or therapeutic treatments. In certain therapeutic applications, an 
adequate amount to accomplish at least partial inhibition, suppression, modulation, 

20 killing, or some other measurable parameter, of a population of selected cells is defined as 
a "therapeutically-effective dose". Amounts needed to achieve this dosage will depend 
upon the severity of the disease and the general state of the patient's own immune system, 
but generally range from 0.005 to 5.0 mg of ligand, e.g. antibody, receptor (e.g. a T-cell 
receptor) or binding protein thereof per kilogram of body weight, with doses of 0.05 to 

25 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions 
containing the present ligands or cocktails thereof may also be administered in similar or 
slightly lower dosages. 

Treatment performed using the compositions described herein is considered "effective" if 
30 one or more symptoms is reduced (e.g., by at least 10% or at least one point on a clinical 
assessment scale), relative to such symptoms present before treatment, or relative to such 
symptoms in an individual (human or model animal) not treated with such composition. 
Symptoms will obviously vary depending upon the disease or disorder targeted, but can 



WO 2004/003019 PCT/GB2003/002804 

80 

be measured by an ordinarily skilled clinician or technician. Such symptoms can be 
measured, for example, by monitoring the level of one or more biochemical indicators of 
the disease or disorder (e.g., levels of an enzyme or metabolite correlated with the 
disease, ' affected cell numbers, etc.), by monitoring physical manifestations (e.g., 
5 inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, 
the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory 
Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect 
to bowel function, systemic symptoms, social function and emotional status - score ranges 
from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life 

10 Rheumatoid Arthritis Scale, or other accepted clinical assessment scale as known in the 
field. A sustained (e.g., one day or more, preferably longer) reduction in disease or 
disorder symptoms by at least 1 0% or by one or more points on a given clinical scale is 
indicative of "effective" treatment. Similarly, prophylaxis performed using a composition 
as described herein is "effective" if the onset or severity of one or more symptoms is 

1 5 delayed, reduced or abolished relative to such symptoms in a similar individual (human or 
animal model) not treated with the composition. 

A composition containing a ligand or cocktail thereof according to the present invention 
may be utilised in prophylactic and therapeutic settings to aid in the alteration, 

20 inactivation, killing or removal of a select target cell population in a mammal. In addition, 
the selected repertoires of polypeptides described herein may be used extracorporeally or 
in vitro selectively to kill, deplete or otherwise effectively remove a target cell population 
from a heterogeneous collection of cells. Blood from a mammal may be combined 
extracorporeally with the ligands, e.g. antibodies, cell-surface receptors or binding 

25 proteins thereof whereby the undesired cells are killed or otherwise removed from the 
blood for return to the mammal in accordance with standard techniques. 



I: Use of half-life enhanced dual-specific ligands according to the invention 

30 

Dual-specific ligands according to the method of the present invention may be employed 
in in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and 
the like. 



WO 2004/003019 



PCT/GB2003/002804 



81 



Therapeutic and prophylactic uses of dual-specific ligands prepared according to the 
invention involve the administration of ligands according to the invention to a recipient 
mammal, such as a human. Dual specific antibodies according to the invention comprise 
5 at least one specificity for a half-life enhancing molecule; one or more further specificities 
may be directed against target molecules. For example, a dual-specific IgG may be 
specific for four epitopes, one of which is on a half-life enhancing molecule. Dual- 
specificity can allow antibodies to bind to multimeric antigen with great avidity. Dual- 
specific antibodies can allow the cross-linking of two antigens, for example in recruiting 
10 cytotoxic T-cells to mediate the killing of tumour cell lines. 

Substantially pure ligands or binding proteins thereof, such as dAb monomers, of at least 
90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or 
more homogeneity is most preferred for pharmaceutical uses, especially when the 
15 mammal is a human. Once purified, partially or to homogeneity as desired, the ligands 
may be used diagnostically or therapeutically (including extracorporeal^) or in 
developing and performing assay procedures, immunofluorescent stainings and the like 
(Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, 
Academic Press, NY). 

20 

The ligands of the present invention will typically find use in preventing, suppressing or 
treating inflammatory states, allergic hypersensitivity, cancer, bacterial or viral infection, 
and autoimmune disorders (which include, but are not limited to, Type I diabetes, 
multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease 
25 and myasthenia gravis). 

In the instant application, the term "prevention" involves administration of the protective 
composition prior to the induction of the disease. "Suppression" refers to administration 
of the composition after an inductive event, but prior to the clinical appearance of the 
30 disease. "Treatment" involves administration of the protective composition after disease 
symptoms become manifest. 



WO 2004/003019 PCT/GB2003/002804 

82 

Animal model systems which can be used to screen the effectiveness of the dual specific 
ligands in protecting against or treating the disease are available. Methods for the testing 
of systemic lupus erythematosus (SLE) in susceptible mice are known in the art (Knight 
et al (1978) J. Exp. Med., 147: 1653; Reinersten et al. (1978) New Eng. J. Med., 299: 
5 515). Myasthenia Gravis (MG) is tested in SJL/J female mice by inducing the disease 
with soluble AchR protein from another species (Lindstrom et al. (1988) Adv. Immunol., 
42: 233). Arthritis is induced in a susceptible strain of mice by injection of Type II 
collagen (Stuart et al. (1984) Ann. Rev. Immunol., 42: 233). A model by which adjuvant 
arthritis is induced in susceptible rats by injection of mycobacterial heat shock protein has 

10 been described (Van Eden et al. (1988) Nature, 331: 171). Thyroiditis is induced in mice 
by administration of thyroglobulin as described (Maron et al. (1980) J. Exp. Med., 152: 
1115). Insulin dependent diabetes mellitus (IDDM) occurs naturally or can be induced in 
certain strains of mice such as those described by Kanasawa et al. (1984) Diabetologia, 
27: 113. EAE in mouse and rat serves as a model for MS in human. In this model, the 

15 demyelinating disease is induced by administration of myelin basic protein (see Paterson 
(1986) Textbook of Immunopathology, Mischer et al., eds., Grune and Stratton, New 
York, pp. 179-213; McFarlin et al. (1973) Science, 179: 478: and Satoh et al. (1987) J. 
Immunol., 138: 179). 

20 Dual specific ligands according to the invention and dAb monomers able to bind to 
extracellular targets involved in endocytosis (e.g. Clathrin) enable dual specific ligands to 
be endocytosed, enabling another specificity able to bind to an intracellular target to be 
delivered to an intracellular environment. This strategy requires a dual specific ligand 
with physical properties that enable it to remain functional inside the cell. Alternatively, 

25 if the final destination intracellular compartment is oxidising, a well folding ligand may 
not need to be disulphide free. 

Generally, the present dual specific ligands will be utilised in purified form together with 
pharmacologically appropriate carriers. Typically, these carriers include aqueous or 
30 alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or 
buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, 
dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable 



WO 2004/003019 PCT/GB2003/002804 

83 

adjuvants, if necessary to keep a polypeptide complex in suspension, may be chosen 
from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and 
alginates. 

5 Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishes, 
such as those based on Ringer's dextrose. Preservatives and other additives, such as 

4 

antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack 
(1982) Remington's Pharmaceutical Sciences, 16th Edition). 

10 The ligands of the present invention may be used as separately administered compositions 
or in conjunction with other agents. These can include various immunotherapeutic drugs, 
such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. 
Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents 
in conjunction with the ligands of the present invention. 

15 

The route of administration of pharmaceutical compositions according to the invention 
may be any of those commonly known to those of ordinary skill in the art. For therapy, 
including without limitation immunotherapy, the ligands of the invention can be 
administered to any patient in accordance with standard techniques. The administration 
20 can be by any appropriate mode, including parenterally, intravenously, intramuscularly, 
intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct 
infusion with a catheter. The dosage and frequency of administration will depend on the 
age, sex and condition of the patient, concurrent administration of other drugs, 
counterindications and other parameters to be taken into account by the clinician. 

25 

The ligands of the invention can be lyophilised for storage and reconstituted in a suitable 
carrier prior to use. This technique has been shown to be effective with conventional 
immunoglobulins and art-known lyophilisation and reconstitution techniques can be 
employed. It will be appreciated by those skilled in the art that lyophilisation and 
30 reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional 
immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) 
and that use levels may have to be adjusted upward to compensate. 



WO 2004/003019 PCT/GB2003/002804 

84 

The compositions containing the present ligands or a cocktail thereof can be 
administered for prophylactic and/or therapeutic treatments. In certain therapeutic 
applications, an adequate amount to accomplish at least partial inhibition, suppression, 
modulation, killing, or some other measurable parameter, of a population of selected cells 
5 is defined as a "therapeutically-effective dose". Amounts needed to achieve this dosage 
will depend upon the severity of the disease and the general state of the patient's own 
immune system, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body 
weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For 
prophylactic applications, compositions containing the present ligands or cocktails thereof 
10 may also be administered in similar or slightly lower dosages. 

A composition containing a ligand according to the present invention may be utilised in 
prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or 
removal of a select target cell population in a mammal. 

15 

In addition, the selected repertoires of polypeptides described herein may be used 
extracorporeal^ or in vitro selectively to kill, deplete or otherwise effectively remove a 
target cell population from a heterogeneous collection of cells. Blood from a mammal 
may be combined extracorporeally with the ligands, e.g. antibodies, cell- surface 
20 receptors or binding proteins thereof whereby the undesired cells are killed or otherwise 
removed from the blood for return to the mammal in accordance with standard 
techniques. 

The invention is further described, for the purposes of illustration only, in the following 
25 examples. As used herein, for the purposes of dAb nomenclature, human TNFa is 
referred to as TAR1 and human TNFa receptor 1 (p55 receptor) is referred to as TAR2. 

Example 1. Selection of a dual specific scFv antibody (K8) directed against human 
30 serum albumin (HSA) and P-galactosidase -gal) 

This example explains a method for making a dual specific antibody directed against 0- 
gal and HSA in which a repertoire of V K variable domains linked to a germline (dummy) 



WO 2004/003019 PCT/GB2003/002804 

85 

Vh domain is selected for binding to p-gal and a repertoire of Vh variable domains 
linked to a germline (dummy) V K domain is selected for binding to HSA. The selected 
variable Vh HSA and V K p-gal domains are then combined and the antibodies selected for 
binding to P-gal and HSA. HSA is a half-life increasing protein found in human blood. 



Four human phage antibody libraries were used in this experiment. 



Library 1 Germline V K /DVT V H 8.46 x 10 7 

Library 2 Germline V K /NNK V H 9.64 x 10 7 

Library 3 Germline V H /DVT V K 1 .47 x 1 0 8 

Library 4 Germline V H /NNK V K 1.45x1 0 8 



All libraries are based on a single human framework for Vh (V3-23/DP47 and J H 4b) and 
V K (012/02/DPK9 and J K 1) with side chain diversity incorporated in complementarity 

determining regions (CDR2 and CDR3). 

Library 1 and Library 2 contain a dummy V K sequence, whereas the sequence of Vh is 

diversified at positions H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97 and H98 
(DVT or NNK encoded, respectively) (Figure 1). Library 3 and Library 4 contain a 
dummy Vh sequence, whereas the sequence of V K is diversified at positions L50, L53, 

L91, L92, L93, L94 and L96 (DVT or NNK encoded, respectively) (Figure 1). The 
libraries are in phagemid pIT2/ScFv format (Figure 2) and have been preselected for 
binding to generic ligands, Protein A and Protein L, so that the majority of clones in the 
unselected libraries are functional. The sizes of the libraries shown above correspond to 
the sizes after preselection. Library 1 and Library 2 were mixed prior to selections on 
antigen to yield a single Vn/duinmy V K library and Library 3 and Library 4 were mixed 

to form a single V K /dummy Vh library. 

Three rounds of selections were performed on P-gal using V K /dummy V H library and 
three rounds of selections were performed on HSA using Vn/diunmy V K library. In the 



WO 2004/003019 PCT/GB2003/002804 

86 

case of p-gal the phage titres went up from 1.1 x 10^ in the first round to 2.0 x 10^ in the 

third round. In the case of HSA the phage titres went up from 2 x l O^ in the first round to 

1.4 x 1()9 in the third round. The selections were performed as described by Griffith et al. 9 
(1993), except that KM13 helper phage (which contains a pill protein with a protease 
5 cleavage site between the D2 and D3 domains) was used and phage were eluted with 1 
mg/ml trypsin in PBS. The addition of trypsin cleaves the pill proteins derived from the 
helper phage (but not those from the phagemid) and elutes bound scFv-phage fusions by 
cleavage in the c-myc tag (Figure 2), thereby providing a further enrichment for phages 
expressing functional scFvs and a corresponding reduction in background (Kristensen & 
10 Winter, Folding & Design 3: 321-328, Jul 9, 1998). Selections were performed using 
immunotubes coated with either HSA or P-gal at lOOjag/ml concentration. 

To check for binding, 24 colonies from the third round of each selection were screened by 
monoclonal phage ELISA. Phage particles were produced as described by Harrison et aL, 
15 Methods Enzymol. 1996;267:83-109. 96-well ELISA plates were coated with 100pl of 
HSA or P-gal at 10|ag/ml concentration in PBS overnight at 4°C. A standard ELISA 
protocol was followed (Hoogenboom et aL, 1991) using detection of bound phage with 
anti-M13-HRP conjugate. A selection of clones gave ELISA signals of greater than 1.0 
with 50|ixl supernatant. 

20 

Next, DNA preps were made from V H /dummy V K library selected on HSA and from 
V K /dummy Vh library selected on p-gal using the QIAprep Spin Miniprep kit (Qiagen). 

To access most of the diversity, DNA preps were made from each of the three rounds of 
selections and then pulled together for each of the antigens. DNA preps were then 
25 digested with SalZ/Not/ overnight at 37°C. Following gel purification of the fragments, 
V K chains from the V K /dummy Vh library selected on p-gal were ligated in place of a 

dummy V K chain of the VH/dummy V K library selected on HSA creating a library of 3.3 
x 10 9 clones. 

30 This library was then either selected on HSA (first round) and P-gal (second round), 
HSA/p-gal selection, or on P-gal (first round) and HSA (second round), p-gal/HSA 



WO 2004/003019 PCT/GB2003/002804 

87 

selection. Selections were performed as described above. Li each case after the second 
round 48 clones were tested for binding to HSA and P-gal by the monoclonal phage 
ELISA (as described above) and by ELISA of the soluble scFv fragments. Soluble 
antibody fragments were produced as described by Harrison et ah, (1996), and standard 
5 ELISA protocol was followed Hoogenboom et al. (1991) Nucleic Acids Res., 19: 4133, 
except that 2% Tween/PBS was used as a blocking buffer and bound scFvs were detected 
with Protein L-HRP. Three clones (E4, E5 and E8) from the HSA/ p-gal selection and two 
clones (K8 and K10) from the p-gal/HSA selection were able to bind both antigens. scFvs 
from these clones were PCR amplified and sequenced as described by Ignatovich et al., 
10 (1999) J Mol Biol 1999 Nov 26;294(2):457-65, using the primers LMB3 and pHENseq. 
Sequence analysis revealed that all clones were identical. Therefore, only one clone 
encoding a dual specific antibody (K8) was chosen for further work (Figure 3). 



15 Example 2. Characterisation of the binding properties of the K8 antibody. 

Firstly, the binding properties of the K8 antibody were characterised by the monoclonal 
phage ELISA. A 9 6 -well plate was coated with 100pl of HSA and P-gal alongside with 
alkaline phosphatase (APS), bovine serum albumin (BSA), peanut agglutinin, lysozyme 

20 and cytochrome c (to check for cross-reactivity) at 10jag/ml concentration in PBS 
overnight at 4°C. The phagemid from K8 clone was rescued with KM13 as described by 
Harrison et aL 9 (1996) and the supernatant (50jli1) containing phage assayed directly. A 
standard ELISA protocol was followed (Hoogenboom et ah, 1991) using detection of 
bound phage with anti-M13-HRP conjugate. The dual specific K8 antibody was found to 

25 bind to HSA and P-gal when displayed on the surface of the phage with absorbance 
signals greater than 1.0 (Figure 4). Strong binding to BSA was also observed (Figure 4). 
Since HSA and BSA are 76% homologous on the amino acid level, it is not surprising 
that K8 antibody recognised both of these structurally related proteins. No cross-reactivity 
with other proteins was detected (Figure 4). 

30 

Secondly, the binding properties of the K8 antibody were tested in a soluble scFv ELISA. 
Production of the soluble scFv fragment was induced by IPTG as described by Harrison 
et al., (1996). To determine the expression levels of K8 scFv, the soluble antibody 



WO 2004/003019 PCT/GB2003/002804 

88 

fragments were purified from the supernatant of 50ml inductions using Protein A- 
Sepharose columns as described by Harlow and Lane, Antibodies: a Laboratory Manual, 
(1988) Cold Spring Harbor. OD28O was ^en measured and the protein concentration 
calculated as described by Sambrook et al. 9 (1989). K8 scFv was produced in supernatant 
5 at 19mg/L 

A soluble scFv ELISA was then performed using known concentrations of the K8 
antibody fragment. A 9 6- well plate was coated with lOOjal of HSA, BSA and p-gal at 
lOjag/ml and lOOpl of Protein A at l|ag/ml concentration. 50|al of the serial dilutions of 
10 the K8 scFv was applied and the bound antibody fragments were detected with Protein L- 
HRP. ELISA results confirmed the dual specific nature of the K8 antibody (Figure 5). 

To confirm that binding to P-gal is determined by the V K domain and binding to 

HSA/BSA by the V H domain of the K8 scFv antibody, the V K domain was cut out from 

15 K8 scFv DNA by SalZ/Not/ digestion and ligated into a SalZ/Not/ digested pIT2 vector 
containing dummy V H chain (Figures 1 and 2). Binding characteristics of the resulting 
clone K8V K /dummy V H were analysed by soluble scFv ELISA. Production of the soluble 

scFv fragments was induced by EPTG as described by Harrison et al. y (1996) and the 
supernatant (50^i) containing scFvs assayed directly. Soluble scFv ELISA was performed 
20 as described in Example 1 and the bound scFvs were detected with Protein L-HRP. The 
ELISA results revealed that this clone was still able to bind P-gal, whereas binding to 
BSA was abolished (Figure 6). 

Example 3. Selection of single Vjj domain antibodies antigens A and B and single 
25 V K domain antibodies directed against antigens C and D. 

This example describes a method for making single Vjj domain antibodies directed 

against antigens A and B and single V K domain antibodies directed against antigens C 

and D by selecting repertoires of virgin single antibody variable domains for binding to 
30 these antigens in the absence of the complementary variable domains. 



WO 2004/003019 PCT/GB2003/002804 

89 

Selections and characterisation of the binding clones is performed as described 
previously (see Example 5, PCT/GB 02/003014). Four clones are chosen for further 
work: 

5 VHl-AntiAV H 

VH2-AntiB Vh 
VK1 - Anti C V K 

VK2~AntiD V K 

10 The procedures described above in Examples 1-3 may be used, in a similar manner as that 
described, to produce dimer molecules comprising combinations of V H domains (i.e., V H - 
V H ligands) and cominations of Vl domains (V l -Vl ligands). 

Example 4. Creation and characterisation of the dual specific ScFv antibodies 
15 (VH1/VH2 directed against antigens A and B and VK1/VK2 directed against 
antigens C and D). 

This example demonstrates that dual specific ScFv antibodies (VH1/VH2 directed against 
antigens A and B and VK1/VK2 directed against antigens C and D) could be created by 
20 combining V K and V H single domains selected against respective antigens in a ScFv 
vector. 

To create dual specific antibody VH1/VH2, VH1 single domain is excised from variable 
domain vector 1 (Figure 7) by NcollXhol digestion and ligated into NcoVXhol digested 
25 variable domain vector 2 (Figure 7) to create VH1/ variable domain vector 2. VH2 single 
domain is PGR amplified from variable domain vector 1 using primers to introduce Sail 
restriction site to the 5' end and Notl restriction site to the 3 5 end. The PGR product is 
then digested with Sail/ Notl and ligated into SaWNotl digested VH1/ variable domain 
vector 2 to create VH1/VH2/ variable domain vector 2. 

30 

VK1 /VK2/ variable domain vector 2 is created in a similar way. The dual specific nature 
of the produced VH1 /VH2 ScFv and VK1/VK2 ScFv is tested in a soluble ScFv ELISA 



WO 2004/003019 PCT/GB2003/002804 

90 

as described previously (see Example 6, PCT/GB 02/003014). Competition ELISA is 
performed as described previously (see Example 8, PCT/GB 02/003014). 



Possible outcomes: 
5 -VH1/VH2 ScFv is able to bind antigens A and B simultaneously 
-VK1/VK2 ScFv is able to bind antigens C and D simultaneously 
-VH1/VH2 ScFv binding is competitive (when bound to antigen A, VH1/VH2 ScFv 
cannot bind to antigen B) 

-VK1/VK2 ScFv binding is competitive (when bound to antigen C, VK1/VK2 ScFv 
10 cannot bind to antigen D) 

Example 5. Construction of dual specific VH1/VH2 Fab and VK1/VK2 Fab and 
analysis of their binding properties, 

15 To create VH1/VH2 Fab, VH1 single domain is ligated into NcoVXfiol digested CH 
vector (Figure 8) to create VH1/CH and VH2 single domain is ligated into SaWNoil 
digested CK vector (Figure 9) to create VH2/CK. Plasmid DNA from VH1/CH and 
VH2/CK is used to co-transform competent E. coli cells as described previously (see 
Example 8, PCT/GB 02/003 01 4). 

20 

The clone containing VH1/CH and VH2/CK plasmids is then induced by IPTG to 
produce soluble VH1/VH2 Fab as described previously (see Example 8, PCT/GB 
02/003014). 



VK1/VK2 Fab is produced in a similar way. 

Binding properties of the produced Fabs are tested by competition ELISA as described 
previously (see Example 8, PCT/GB 02/003014). 



30 



Possible outcomes: 

-VH1/VH2 Fab is able to bind antigens A and B simultaneously 
-VK1/VK2 Fab is able to bind antigens C and D simultaneously 



WO 2004/003019 PCT/GB2003/002804 

91 

-VH1/VH2 Fab binding is competitive (when bound to antigen A 5 VH1/VH2 Fab cannot 
bind to antigen B) 

-VK1/VK2 Fab binding is competitive (when bound to antigen C, VK1/VK2 Fab cannot 
bind to antigen D) 

5 

Example 6 

Chelating dAb Dimers 
10 Summary 

VH and VK homo-dimers are created in a dAb-linker~dAb format using flexible 
polypeptide linkers. Vectors were created in the dAb linker-dAb format containing 
glycine-serine linkers of different lengths 3U:(Gly4Ser) 3 , 5U:(Gly4Ser)5 ? 7U:(Gly4Ser)7. 
Dimer libraries were created using guiding dAbs upstream of the linker: TAR1-5 (VK) 5 
15 TAR1-27(VK) 3 TAR2-5(VH) or TAR2-6(VK) and a library of corresponding second 
dAbs after the linker. Using this method, novel dimeric dAbs were selected. The effect of 
dimerisation on antigen binding was determined by ELISA and BIAcore studies and in 
cell neutralisation and receptor binding assays. Dimerisation of both TAR1-5 and TAR1- 
27 resulted in significant improvement in binding affinity and neutralisation levels. 

20 

1.0 Methods 

1.1 Library generation 
1.1.1 Vectors 

pEDASU, pEDA5U and pEDA7U vectors were designed to introduce different linker 
25 lengths compatible with the dAb - linker- dAb format. For pEDA3U, sense and anti-sense 
73-base pair oligo linkers were annealed using a slow annealing program (95°C-5mins, 
80°C-10mins, 70°C-15mins, 56°C-15mins ? 42°C until use) in buffer containing 
O.lMNaCl, lOmM Tris-HCl pH7.4 and cloned using the Xltol and Notl restriction sites. 
The linkers encompassed 3 (Gly4Ser) units and a stuffer region housed between Sail and 
30 Notl cloning sites (scheme 1). In order to reduce the possibility of monomelic dAbs 
being selected for by phage display, the stuffer region was designed to include 3 stop 
codons, a Sacl restriction site and a frame shift mutation to put the region out of frame 
when no second dAb was present. For pEDASU and 7U due to the length of the linkers 



WO 2004/003019 PCT/GB2003/002804 

92 

required, overlapping oligo -linkers were designed for each vector, annealed and 
elongated using Klenow. The fragment was then purified and digested using the 
appropriate enzymes before cloning using tho Xhol and Notl restriction sites. 



Ncol 



I 



Xhol 



Stuffer 1 



Linker: 
3U 



5U 



Sail 



7U 



1 



Notl 



Stuffer 2 



Scheme 1 



1.1.2 Library preparation 

The N-terminal V gene corresponding to the guiding dAb was cloned upstream of the 
10 linker using Ncol and Xhol restriction sites. VH genes have existing compatible sites, 
however cloning VK genes required the introduction of suitable restriction sites. This was 
achieved by using modifying PCR primers (VK-DLIBF: 5' cggccatggcgtcaacggacat; 
VKXholR: 5' atgtgcgctcgagcgtttgattt 3') in 30 cycles of PCR amplification using a 2:1 
mixture of SuperTaq (HTBiotechnology Ltd)and pfu turbo (Stratagene). This maintained 
15 the Ncol site at the 5' end while destroying the adjacent Sail site and introduced the 
Xhol site at the 3' end. 5 guiding dAbs were cloned into each of the 3 dimer vectors: 
TAR1-5 (VK), TAR1-27(VK), TAR2-5(VH), TAR2-6(VK) and TAR2-7(VK). All 
constructs were verified by sequence analysis. 



20 Having cloned the guiding dAbs upstream of the linker in each of the vectors (pEDA3U, 
5U and 7U): TAR1-5 (VK), TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) a library of 
corresponding second dAbs were cloned after the linker. To achieve this, the 
complimentary dAb libraries were PCR amplified from phage recovered from round 1 
selections of either a VK library against Human TNFa (at approximately 1 x 10 6 diversity 

25 after round 1) when TAR 1-5 or TAR1-27 are the guiding dAbs, or a VH or VK library 
against human p55 TNF receptor (both at approximately 1 x 10 5 diversity after round 1) 
when TAR2-5 or TAR2-6 respectively are the guiding dAbs. For VK libraries PCR 
amplification was conducted using primers in 30 cycles of PCR amplification using a 2:1 
mixture of SuperTaq and pfu turbo. VH libraries were PCR amplified using primers in 



WO 2004/003019 PCT/GB2003/002804 

93 

order to introduce a Sail restriction site at the 5' end of the gene. The dAb library PCRs 
were digested with the appropriate restriction enzymes, ligated into the corresponding 
vectors down stream of the linker, using Sall/Notl restriction sites and electroporated 
into freshly prepared competent TGI cells. 

5 

The titres achieved for each library are as follows: 
TAR1-5: pEDA3U = 4xl0 8 , pEDASU = 8xl0 7 , pEDA7U = 1x10 s 
TAR1-27: pEDASU = 6.2xl0 8 , pEDASU - 1x10 s , pEDA7U = lxlO 9 
TAR2h-5 : pEDA3U = 4xl0 7 , pEDA5U - 2 x 10 8 , pEDA7U = 8xl0 7 
10 TAR2h-6: pEDA3U = 7.4xl0 8 5 pEDASU = 1.2 x 10 8 , pEDA7U = 2.2xl0 8 



1.2 Selections 
1.2.1 TNFa 

Selections were conducted using human TNFa passively coated on immunotubes. 

15 Briefly, Immunotubes are coated overnight with l-4mls of the required antigen. The 
immunotubes were then washed 3 times with PBS and blocked with 2%milk powder in 
PBS for l~2hrs and washed a further 3 times with PBS. The phage solution is diluted in 
2%milk powder in PBS and incubated at room temperature for 2hrs. The tubes are then 
washed with PBS and the phage eluted with lmg/ml trypsin-PBS. Three selection 

20 strategies were investigated for the TAR1-5 dimer libraries. The first round selections 
were carried out in immunotubes using human TNFa coated at ljixg/ml or 20|Lig/ml with 
20 washes in PBS 0.1%Tween. TGI cells are infected with the eluted phage and the titres 
are determined (eg, Marks et al J Mol Biol. 1991 Dec 5;222(3):581-97, Richmann et al 
Biochemistry. 1993 Aug 31;32(34):8848-55). 

25 

The titres recovered were: 

pEDA3U = 2.8xl0 7 (ljig/ml TNF) 1.5xl0 8 (20|ag/mlTNF), 
pEDASU = 1.8xl0 7 (l|ng/ml TNF), 1.6xl0 8 (20jag/ml TNF) 
pEDA7U = 8xl0 6 (ljLig/ml TNF), 7xl0 7 (20jag/ml TNF). 

30 

The second round selections were carried out using 3 different methods. 

1. In immunotubes, 20 washes with overnight incubation followed by a further 10 
washes. 



WO 2004/003019 PCT/GB2003/002804 

94 

2. In immunotubes, 20 washes followed by lhr incubation at RT in wash buffer 

with (lp,g/ml TNFa) and 10 further washes. 
3. Selection on streptavidin beads using 33 pmoles biotinylated human TNFa 
(Henderikx et aL 9 2002, Selection of antibodies against biotinylated antigens. 
Antibody Phage Display : Methods and protocols, Ed. O'Brien and Atkin, Humana 
Press). Single clones from round 2 selections were picked into 96 well plates and 
crude supernatant preps were made in 2ml 96 well plate format. 





Round 1 


Round 2 


Round 2 


Round 2 




Human 


selection 


selection 


selection 




TNFaimmuno 


method 1 


method 2 


method 3 




tube coating 
concentration 








pEDA3U 


l|_ig/ml 


1 x 10 y 


1.8 x 10 y 


2.4 x 10 10 


pEDA3U 


20p,g/ml 


6x 10 y 


1.8 x 10 10 


8.5 x 10 10 


pEDA5U 


l|j.g/ml 


9x 10 a 


1.4 x 10 y 


2.8 x 10 10 


pEDA5U 


20u.g/ml 


9.5 x 10 y 


8.5 x 10 y 


2.8 x 10 10 


pEDA7U 


l(j.g/ml 


7.8 x 10* 


1.6 x 10 8 


4x 10 10 


pEDA7U 


20u.g/ml 


1 x 10 10 


8x 10 y 


1.5 x 10 10 



For TAR 1-27, selections were carried out as described previously with the following 
modifications. The first round selections were carried out in immunotubes using human 
TNFa coated at ljag/ml or 20|ug/ml with 20 washes in PBS 0.1%Tween. The second 
round selections were carried out in immunotubes using 20 washes with overnight 
incubation followed by a further 20 washes. Single clones from round 2 selections were 
picked into 96 well plates and crude supernatant preps were made in 2ml 96 well plate 
format. 



TAR 1-2 7 titres are as follows 



WO 2004/003019 



95 



PCT/GB2003/002804 





Human 

TNFaimmunotu.be 
coating cone 


Round 1 


Round 2 


pEDA3U 


lug/ml 


4x 10 y 


6x 10 y 


pEDA3U 


20ug/ml 


5x 10 y 


4.4 x 10 1U 


pEDA5U 


lug/ml 


1.5 x 10 9 


1.9 x 10 1U 


pEDA5U 


20ug/ml 


3.4 x 10 y 


3.5 x 10 1U 


pEDA7U 


lug/ml 


2.6 x 10 y 


5 x 10 y 


pEDA7U 


20(j.g/ml 


7x 10 y 


1.4 x 10 1U 



1.2.2 TNF RECEPTOR 1 (p55 RECEPTOR; TAR2) 

Selections were conducted as described previously for the TAR2h-5 libraries only. 3 
rounds of selections were carried out in immunotubes using either ljag/ml human p55 
TNF receptor or 10jng/ml human p55 TNF receptor with 20 washes in PBS 0.1%Tween 
with overnight incubation followed by a further 20 washes. Single clones from round 2 
and 3 selections were picked into 96 well plates and crude supernatant preps were made 
in 2ml 96 well plate format. 

TAR2h-5 titres are as follows: 





Round 1 human 

p55TNF 

receptor 

immunotube 

coating 

concentration 


Round 1 


Round 2 


Round 3 


pEDA3U 


lu,g/ml 


2.4 x 10 6 


1.2 x 10' 


1.9 x 10 y 


pEDA3U 


lOug/ml 


3.1 x 10 7 


7 x 10 7 


1 x 10 y 


pEDA5U 


lug/ml 


2.5 x 10 6 


1.1 x 10 7 


5.7 x 10 s 


pEDA5U 


lOug/ml 


3.7 x 10 7 


2.3 x 10 s 


2.9 x 10 y 


pEDA7U 


lug/ml 


1.3 x 10 6 


1.3 x 10 7 


1.4 x 10 y 


pEDA7U 


10|j,g/ml 


1.6 x 10 7 


1.9 x 10 7 


3 x 10 1U 



1.3 Screening 

Single clones from round 2 or 3 selections were picked from each of the 3U, 5U and 7U 
libraries from the different selections methods, where appropriate. Clones were grown in 
2xTY with lOOjag/ml ampicillin and 1% glucose overnight at 37°C. A 1/100 dilution of 
this culture was inoculated into 2mls of 2xTY with 100|ng/ml ampicillin and 0.1% 
glucose in 2ml, 96 well plate format and grown at 37°C shaking until OD600 was 



WO 2004/003019 PCT/GB2003/002804 

96 

approximately 0.9. The culture was then induced with ImM IPTG overnight at 30°C. 
The supernatants were clarified by centrifugation at 4000rpm for 15 rnins in a sorval plate 
centrifuge. The supernatant preps the used for initial screening. 

5 1.3.1 ELISA 

Binding activity of dimeric recombinant proteins was compared to monomer by Protein 
A/L ELISA or by antigen ELISA. Briefly, a 96 well plate is coated with antigen or 
Protein A/L overnight at 4°C. The plate washed with 0.05% Tween-PBS, blocked for 2hrs 
with 2% Tween-PBS. The sample is added to the plate incubated for 1 hr at room 

10 temperature. The plate is washed and incubated with the secondary reagent for lhr at 
room temperature. The plate is washed and developed with TMB substrate. Protein A/L- 
HRP or India-HRP was used as a secondary reagent. For antigen ELISAs, the antigen 
concentrations used were l|ug/ml in PBS for Human TNFa and human THF receptor 1. 
Due to the presence of the guiding dAb in most cases dimers gave a positive ELISA 

15 signal therefore off rate determination was examined by BIAcore. 

1.3.2 BIAcore 

BIAcore analysys was conducted for TAR1-5 and TAR2h-5 clones. For screening, 
Human TNFawas coupled to a CMS chip at high density (approximately 10000 RUs). 
20 50 jlxI of Human TNFoc(50 jag/ml) was coupled to the chip at 5jal/min in acetate buffer - 
pH5.5. Regeneration of the chip following analysis using the standard methods is not 
possible due to the instability of Human TNFa, therefore after each sample was analysed, 
the chip was washed for 1 Omins with buffer. 

For TAR1-5, clones supernatants from the round 2 selection were screened by BIAcore. 
25 48 clones were screened from each of the 3U, 5U and 7U libraries obtained using the 
following selection methods: 

Rl: 1/xg/ml human TNFa immunotube, R2 1 /xg/ml human TNFa immunotube, overnight 
wash. 

Rl: 20/xg/ml human TNFa immunotube, R2 20/xg/ml human TNFa immunotube, 
30 overnight wash. 

Rl: 1/xg/ml human TNFa immunotube, R2 33 pmoles biotinylated human TNFa on 
beads. 

Rl: 20/xg/ml human TNFa immunotube, R2 33 pmoles biotinylated human TNFa beads. 



WO 2004/003019 



97 



PCT/GB2003/002804 



For screening, human p55 TNF receptor was coupled to a CMS chip at high density 
(approximately 4000 RUs). 100 jlxI of human p55 TNF receptor (10 (ag/ml) was coupled 
to the chip at 5jjl/min in acetate buffer - pH5.5. Standard regeneration conditions were 
5 examined ( glycine pH2 or pH3) but in each case antigen was removed from the surface 
of the chip therefore as with TNFa, therefore after each sample was analysed, the chip 
was washed for 1 Omins with buffer. 

For TAR2-5, clones supernatants from the round 2 selection were screened. 
48 clones were screened from each of the 3U, 5U and 7U libraries, using the following 
1 o selection methods : 

Rl: 1/zg/ml human p55 TNF receptor immunotube, R2 1/xg/ml human p55 TNF receptor 
immunotube, overnight wash. 

Rl: 10/xg/ml human p55 TNF receptor immunotube, R2 1 0/xg/ml human p55 TNF 
receptor immunotube, overnight wash. 

15 

1.3.3 Receptor and Cell Assays 

The ability of the dimers to neutralise in the receptor assay was conducted as follows: 
Receptor binding 

20 Anti-TNF dAbs were tested for the ability to inhibit the binding of TNF to recombinant 
TNF receptor 1 (p55). Briefly, Maxisorp plates were incubated overnight with 30mg/ml 
anti-human Fc mouse monoclonal antibody (Zymed, San Francisco, USA). The wells 
were washed with phosphate buffered saline (PBS) containing 0.05% Tween-20 and then 
blocked with 1% BS A in PBS before being incubated with lOOng/ml TNF receptor 1 Fc 

25 fusion protein (R&D Systems, Minneapolis, USA). Anti-TNF dAb was mixed with TNF 
which was added to the washed wells at a final concentration of lOng/ml. TNF binding 
was detected with 0.2mg/ml biotinylated anti-TNF antibody (HyCult biotechnology, 
Uben, Netherlands) followed by 1 in 500 dilution of horse radish peroxidase labelled 
streptavidin (Amersham Biosciences, UK) and then incubation with TMB substrate (KPL, 

30 Gaithersburg, USA). The reaction was stopped by the addition of HC1 and the absorbance 
was read at 450nm. Anti-TNF dAb activity lead to a decrease in TNF binding and 
therefore a decrease in absorbance compared with the TNF only control. 



WO 2004/003019 PCT/GB2003/002804 

98 

L929 Cytotoxicity Assay 

Anti-TNF dAbs were also tested for the ability to neutralise the cytotoxic activity of TNF 
on mouse L929 fibroblasts (Evans, T. (2000) Molecular Biotechnology 15, 243-248). 
Briefly, L929 cells plated in microtitre plates were incubated overnight with anti-TNF 
5 dAb, lOOpg/ml TNF and lmg/ml actinomycin D (Sigma, Poole, UK). Cell viability was 
measured by reading absorbance at 490nm following an incubation with [3-(4,5- 
dimethylthiazol-2-yl)-5-(3-carbboxym^ 

(Promega, Madison, USA). Anti-TNF dAb activity lead to a decrease in TNF cytotoxicity 
and therefore an increase in absorbance compared with the TNF only control. 

10 

In the initial screen, supernatants prepared for BIAcore analysis, described above, were 
also used in the receptor assay. Further analysis of selected dimers was also conducted in 
the receptor and cell assays using purified proteins. 

15 HeLa IL-8 assay 

Anti-TNFRl or anti-TNF alpha dAbs were tested for the ability to neutralise the 
induction of IL-8 secretion by TNF in HeLa cells (method adapted from that of Akeson, 
L. et al (1996) Journal of Biological Chemistry 271, 30517-30523, describing the 
induction of IL-8 by IL-1 in HUVEC; here we look at induction by human TNF alpha and 
20 we use HeLa cells instead of the HUVEC cell line). Briefly, HeLa cells plated in 
microtitre plates were incubated overnight with dAb and 300pg/ml TNF. Post incubation 
the supernatant was aspirated off the cells and IL-8 concentration measured via a 
sandwich ELISA (R&D Systems). Anti-TNFRl dAb activity lead to a decrease in IL-8 
secretion into the supernatant compared with the TNF only control. 

25 

The L929 assay is used throughout the following experiments; however, the use of the 
HeLa IL-8 assay is preferred to measure anti-TNF receptor 1 (p55) ligands; the presence 
of mouse p55 in the L929 assay poses certain limitations in its use. 

30 1.4 Sequence analysis 

Dimers that proved to have interesting properties in the BIAcore and the receptor assay 
screens were sequenced. Sequences are detailed in the sequence listing. 



WO 2004/003019 PCT/GB2003/002804 

99 

1.5 Formatting 

1.5.1 TAR1-5-19 dimers 

The TAR1-5 dimers that were shown to have good neutralisation properties were re- 
formatted and analysed in the cell and receptor assays. The TAR1-5 guiding dab was 
5 substituted with the affinity matured clone TAR1-5-19. To achieve this TAR1-5 was 
cloned out of the individual dimer pair and substituted with TAR1-5-19 that had been 
amplified by PCR. In addition, TAR1-5-19 homodimers were also constructed in the 3U, 
5U and 7U vectors. The N terminal copy of the gene was amplified by PCR and cloned as 
described above and the C-terminal gene fragment was cloned using existing Sail and 
10 Notl restriction sites. 

1.5.2 Mutagenesis 

The amber stop codon present in dAb2, one of the C-terminal dAbs in the TAR1-5 dimer 
pairs was mutated to a glutamine by site-directed mutagenesis. 

15 

1.5.3 Fabs 

The dimers containing TAR1-5 or TAR 1-5- 19 were re-formatted into Fab expression 
vectors. dAbs were cloned into expression vectors containing either the CK or CH genes 
using Sfil and Notl restriction sites and verified by sequence analysis. The CK vector is 
20 derived from a pUC based ampicillin resistant vector and the CH vector is derived from a 
pACYC chloramphenicol resistant vector. For Fab expression the dAb-CH and dAb-CK 
constructs were co-transformed into HB2151 cells and grown in 2xTY containing 0.1% 
glucose, 100jj,g/ml ampicillin and 10/xg/ml chloramphenicol. 

25 1.5.3 Hinge dimerisation 

Dimerisation of dAbs via cystine bond formation was examined. A short sequence of 
amino acids EPKSGDKTHTCPPCP a modified form of the human IgGCl hinge was 
engineered at the C terminal region on the dAb. An oligo linker encoding for this 
sequence was synthesised and annealed, as described previously. The linker was cloned 
30 into the pEDA vector containing TAR1-5-19 using Xliol and Notl restriction sites. 
Dimerisation occurs in situ in the periplasm. 



1.6 Expression and purification 



WO 2004/003019 PCT/GB2003/002804 

100 

1.6.1 Expression 

Supernatants were prepared in the 2ml ? 96-well plate format for the initial screening as 
described previously. Following the initial screening process selected dimers were 
analysed further. Dimer constructs were expressed in TOPI OF' or HB2151 cells as 
5 supernatants. Briefly, an individual colony from a freshly streaked plate was grown 
overnight at 37°C in 2xTY with lOO^ig/ml ampicillin and 1% glucose. A 1/100 dilution 
of this culture was inoculated into 2xTY with 100p,g/ml ampicillin and 0.1% glucose and 
grown at 37°C shaking until OD600 was approximately 0.9. The culture was then induced 
with ImM EPTG overnight at 30°C. The cells were removed by centrifugation and the 

10 supernatant purified with protein A or L agarose. 

Fab and cysteine hinge dimers were expressed as periplasmic proteins in HB2152 cells. 
A 1/100 dilution of an overnight culture was inoculated into 2xTY with 0.1% glucose 
and the appropriate antibiotics and grown at 30°C shaking until OD600 was 
approximately 0.9. The culture was then induced with ImM IPTG for 3-4 hours at 25°C. 

15 The cells were harvested by centrifugation and the pellet resuspended in periplasmic 
preparation buffer (30mM Tris-HCl pH8.0, ImM EDTA, 20% sucrose). Following 
centrifugation the supernatant was retained and the pellet resuspended in 5mM MgS04. 
The supernatant was harvested again by centrifugation, pooled and purified. 

20 1.6.2 Protein A/L purification 

Optimisation of the purification of dimer proteins from Protein L agarose (Affitech, 
Norway) or Protein A agarose (Sigma, UK) was examined. Protein was eluted by batch or 
by column elution using a peristaltic pump. Three buffers were examined 0.1M 
Phosphate-citrate buffer pH2.6, 0.2M Glycine pH2.5 and 0.1M Glycine pH2.5. The 
25 optimal condition was determined to be under peristaltic pump conditions using 0.1M 
Glycine pH2.5 over 10 column volumes. Purification from protein A was conducted 
peristaltic pump conditions using 0.1M Glycine pH2.5. 

1.6.3 FPLC purification 

30 Further purification was carried out by FPLC analysis on the AKTA Explorer 100 system 
(Amersham Biosciences Ltd). TAR1-5 and TAR1-5-19 dimers were fractionated by 
cation exchange chromatography (1ml Resource S - Amersham Biosciences Ltd) eluted 
with a 0-1M NaCl gradient in 50mM acetate buffer pH4. Hinge dimers were purified by 



WO 2004/003019 PCT/GB2003/002804 

101 

ion exchange (1ml Resource Q Amersham Biosciences Ltd) eluted with a 0-1M NaCl 
gradient in 25mMTris HC1 pH 8.0. Fabs were purified by size exclusion chromatography 
using a superose 12 (Amersham Biosciences Ltd ) column run at a flow rate of 0.5ml/min 
in PBS with 0.05% tween. Following purification samples were concentrated using 
5 vivaspin 5K cut off concentrators (Vivascience Ltd). 

2.0 Results 

2.1 TAR1-5 dimers 

6 x 96 clones were picked from the round 2 selection encompassing all the libraries and 
selection conditions. Supernatant preps were made and assayed by antigen and Protein L 
ELISA, BIAcore and in the receptor assays. In ELISAs, positive binding clones were 
identified from each selection method and were distributed between 3U, 5U and 7U 
libraries. However, as the guiding dAb is always present it was not possible to 
discriminate between high and low affinity binders by this method therefore BIAcore 
analysis was conducted. 

BIAcore analysis was conducted using the 2ml sup ernat ants. BIAcore analysis revealed 
that the dimer Koff rates were vastly improved compared to monomelic TAR 1-5. 
Monomer Koff rate was in the range of lO^M compared with dimer Koff rates which 
- 20 were in the range of 10" 3 - 10" 4 M. 16 clones that appeared to have very slow off rates 
were selected, these came from the 3U, 5U and 7U libraries and were sequenced. In 
addition the supernatants were analysed for the ability to neutralise human TNFa in the 
receptor assay. 

25 6 lead clones (dl-d6 below) that neutralised in these assays and have been sequenced. 
The results shows that out of the 6 clones obtained there are only 3 different second dAbs 
(dAb 1 , dAb2 and dAb3) however where the second dAb is found more than once they are 
linked with different length linkers. 

30 TARl-5dl: 3U linker 2 nd &Ab=dAbl - ljj,g/ml Ag immunotube overnight wash 
TARl-5d2: 3U linker 2 nd dAb=dAb2 - lfig/ml Ag immunotube overnight wash 
TARl-5d3: 5U linker 2 nd dAb=dAb2 - ljag/ml Ag immunotube overnight wash 
TARl-5d4: 5U linker 2 nd <±Ab=dAb3 - 20|ug/ml Ag immunotube overnight wash 



10 



15 



WO 2004/003019 PCT/GB2003/002804 

102 

TARl~5d5: 5U linker 2 nd d Ab=d Ab 1 - 20|ag/ml Ag immunotube overnight wash 
TARl-5d6: 7U linker 2 nd dAb=dAbl- Rl:l|j,g/ml Ag immunotube overnight wash, 
R2:beads 

The 6 lead clones were examined further. Protein was produced from the periplasm and 
supernatant, purified with protein L agarose and examined in the cell and receptor assays. 
The levels of neutralisation were variable (Table 1). The optimal conditions for protein 
preparation were determined. Protein produced from HB2151 cells as supematants gave 
the highest yield (approximately lOmgs/L of culture). The supematants were incubated 
with protein L agarose for 2hrs at room temperature or overnight at 4°C. The beads were 
washed with PBS/NaCl and packed onto an FPLC column using a peristaltic pump. The 
beads were washed with 10 column volumes of PBS/NaCl and eluted with 0.1M glycine 
pH2.5. In general, dimeric protein is eluted after the monomer. 

TARl-5dl-6 dimers were purified by FPLC. Three species were obtained, by FPLC 
purification and were identified by SDS PAGE. One species corresponds to monomer and 
the other two species corresponds to dimers of different sizes. The larger of the two 
species is possibly due to the presence of C terminal tags. These proteins were examined 
in the receptor assay. The data presented in table 1 represents the optimum results 
obtained from the two dimeric species (Figure 1 1) 

The three second dAbs from the dimer pairs (ie, dAbl, dAb2 and dAb3) were cloned as 
monomers and examined by ELISA and in the cell and receptor assay. All three dAbs 
bind specifically to TNF by antigen ELISA and do not cross react with plastic or BSA. As 
monomers, none of the dAbs neutralise in the cell or receptor assays. 

2.1.2 TAR1-5-19 dimers 

TAR1-5-19 was substituted for TAR1-5 in the 6 lead clones. Analysis of all TAR1-5-19 
dimers in the cell and receptor assays was conducted using total protein (protein L 
purified only) unless otherwise stated (Table 2). TARl-5-19d4 and TARl-5-19d3 have 
the best ND50 (~5nM) in the cell assay, this is consistent with the receptor assay results 
and is an improvement over TAJR1-5-19 monomer (ND 5 o~30nM). Although purified 
TAR1-5 dimers give variable results in the receptor and cell assays TAR1-5-19 dimers 



WO 2004/003019 PCT/GB2003/002804 

103 

were more consistent. Variability was shown when using different elution buffers during 
the protein purification. Elution using 0.1M Phosphate-citrate buffer pH2.6 or 0.2M 
Glycine pH2.5 although removing all protein from the protein L agarose in most cases 
rendered it less functional. 

5 

TARl-5-19d4 was expressed in the fermenter and purified on cation exchange FPLC to 
yield a completely pure dimer. As with TARl-5d4 three species were obtained, by FPLC 
purification corresponding to monomer and two dimer species. This dimer was amino 
acid sequenced. TAR1-5-19 monomer and TARl-5-19d4 were then examined in the 
10 receptor assay and the resulting IC50 for monomer was 30nM and for dimer was 8nM. 
The results of the receptor assay comparing TAR1-5-19 monomer, TARl-5-19d4 and 
TARl-5d4 is shown in figure 10. 



TAR1-5-19 homodimers were made in the 3U, 5U and 7U vectors, expressed and purified 
15 on Protein L. The proteins were examined in the cell and receptor assays and the resulting 
IC 50 s (for receptor assay) and ND 50 s (for cell assay) were determined (table 3, figure 12). 



2.2 Fabs 

TAR1-5 and TAR1-5-19 dimers were also cloned into Fab format, expressed and purified 
20 on protein L agarose. Fabs were assessed in the receptor assays (Table 4). The results 
showed that for both TAR1-5-19 and TAR1-5 dimers the neutralisation levels were 
similar to the original Gly 4 Ser linker dimers from which they were derived. A TAR1-5-19 
Fab where TAR1-5-19 was displayed on both CH and CK was expressed, protein L 
purified and assessed in the receptor assay. The resulting IC50 was approximately InM. 



2.3 TAR1-27 dimers 

3 x 96 clones were picked from the round 2 selection encompassing all the libraries and 
selection conditions. 2ml supernatant preps were made for analysis in ELISA and 
30 bioassays. Antigen ELISA gave 71 positive clones. The receptor assay of crude 
supernatants yielded 42 clones with inhibitory properties (TNF binding 0-60%). In the 
majority of cases inhibitory properties correlated with a strong ELISA signal. 42 clones 



WO 2004/003019 PCT/GB2003/002804 

104 

were sequenced, 39 of these have unique second dAb sequences. The 12 dimers that 
gave the best inhibitory properties were analysed further. 

The 12 neutralising clones were expressed as 200ml supernatant preps and purified on 
protein L. These were assessed by protein L and antigen ELISA, BIAcore and in the 
receptor assay. Strong positive ELISA signals were obtained in all cases. BIAcore 
analysis revealed all clones to have fast on and off rates. The off rates were improved 
compared to monomelic TAR1-27, however the off rate of TAR1-27 dimers was faster 
(Koff is approximately in the range of 10" 1 and 10" 2 M) than the TAR1-5 dimers examined 
previously (Koff is approximately in the range of 10~ 3 - 10" 4 M). The stability of the 
purified dimers was questioned and therefore in order to improve stability, the addition on 
5%glycerol, 0.5% Triton X100 or 0.5% NP40 (Sigma) was included in the purification of 

2 TAR1-27 dimers (d2 and dl6). Addition of NP40 or Triton X100™ improved the yield 
of purified product approximately 2 fold. Both dimers were assessed in the receptor 
assay. TARl-27d2 gave IC50 of ~30nM under all purification conditions. TARl-27dl6 
showed no neutralisation effect when purified without the use of stabilising agents but 
gave an IC50 of ~50nM when purified under stabilising conditions. No further analysis 
was conducted. 

2.4 TAR2-5 dimers 

3 x 96 clones were picked from the second round selections encompassing all the libraries 
and selection conditions. 2ml supernatant preps were made for analysis. Protein A and 
antigen ELISAs were conducted for each plate. 30 interesting clones were identified as 
having good off-rates by BIAcore (Koff ranges between 10' 2 - 10" 3 M). The clones were 
sequenced and 13 unique dimers were identified by sequence analysis. 



Table 1: TAR1-5 dimers 



Dimer 


Cell 
type 


Purification 


Protein 
Fraction 


Elution 
conditions 


Receptor/ 
Cell assay 


TARl-5dl 


HB2151 


Protein L + 
FPLC 


small dimeric 
species 


0.1M glycine 
pH2.5 


RA~30nM 


TARl-5d2 


HB2151 


Protein L + 
FPLC 


small dimeric 
species 


0.1M glycine 
pH2.5 


RA~50nM 



WO 2004/003019 



105 



PCT/GB2003/002804 







FPLC 


species 


pH2.5 


M 


TARl-5d3 


HB2151 


Protein L + 
FPLC 


large dimeric 
species 


0.1M glycine 
pH2.5 


RA-300 
nM 


TARl-5d4 


HB2151 


Protein L + 
FPLC 


small dimeric 
species 


0.1M glycine 
pH2.5 


RA~3n 
M 


TARl-5d5 


HB2151 


Protein L + 
FPLC 


large dimeric 
species 


0.1M glycine 
pH2.5 


RA-200 
nM 


TARl-5d6 


HB2151 


Protein L 
+FPLC 


Large dimeric 
species 


0.1M glycine 
pH2.5 


RA-100 
nM 



*note dimer 2 and dimer 3 have the same second dAb (called dAb2), however have 
different linker lengths (d2 = (Gly 4 Ser) 3 , d3 - (Gly 4 Ser) 3 ). dAbl is the partner dAb to 
dimers 1, 5 and 6. dAb3 is the partner dAb to dimer4. None of the partner dAbs 
5 neutralise alone. FPLC purification is by cation exchange unless otherwise stated. The 

optimal dimeric species for each dimer obtained by FPLC was determined in these 
assays. 



10 Table 2: TAR1-5-19 dimers 



Dimer 


Cell type 


Purification 


Protein 
Fraction 


Elution conditions 


Recept 
or/ Cell 

assay 


TARl-5-19dl 


TOPI OF* 


Protein L 


Total protein 


0.1M glycine pH 2.0 


RA-15 
nM 


TAR1-5-19 d2 (no 
stop codon) 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH 2.0 + 
0.05%NP40 


RA~2n 
M 


TARl-5-19d3 
(no stop codon) 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH 2.5.+ 
0.05%NP40 


RA~8n 
M 


TARl-5-19d4 


TOPI OF' 


Protein L + 
FPLC 


FPLC purified 
fraction 


0.1M glycine 
pH2.0 


RA~2- 
5nM 
CA-12 
nM 


TARl-5-19d5 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH2.0 + 
NP40 


RA~8n 
M 

CA-10 
nM 


TARl-5-19d6 


TOPI OF 5 


Protein L 


Total protein 


0.1M glycine pH 2.0 


RA-10 
nM 



Table 3: TAR1-5-19 homodimers 



Dimer 


Cell type 


Purification 


Protein Fraction 


Elution conditions 


Recept 










or/ Cell 












assay 



WO 2004/003019 



,106 



PCT/GB2003/002804 



homodimer 










nM 

CA-30 
nM 


TAR1-5-19 5U 
homodimer 


HB2151 


Protein L 


Total protein 


0.1M glycine pH2.5 


RA~2n 
M 

M 


TAR 1-5- 19 7U 
homodimer 


HB2151 




1 ULdl piULClll 


u.iivi glycine priz.3 


nM 

CA-15 
nM 


TAR1-5-19 cys 
hinge 


HB2151 


Protein L + FPLC 


FPLC • purified 
dimer fraction 


0.1M glycine pH2.5 


RA~2n 
M 


TAR1-5-19CH/ 
TAR1-5-19 CK 


HB2151 


Protein 


Total protein 


0.1M glycine pH2.5 


RA~ln 
M 


Table 4: TAR1-5/TAR1-5-19 Fabs 


Dimer 


Cell 

type 


Purification 


Protein 
Fraction 


Elution 
conditions 


Rece 
ptor/ 
Cell 

assay 


TAR1-5CH/ 
dAbl CK 


HB2151 


Protein L 


Total protein 


0,1M citrate pH2.6 


RA-90 
nM 


TAR1-5CH/ 
dAb2 CK 


HB2151 


Protein L 


Total protein 


(MM glycine pH2.5 


RA-30 
nM 

CA-60 
nM 


dAb3CH/ 
TAR1-5CK 


HB2151 


Protein L 


Total protein 


0.1M citrate pH2.6 


RA-10 
OnM 


TAR1-5-19CH/ 
dAbl CK 


HB2151 


Protein L 


Total protein 


0.1M glycine pH2.0 


RA~6n 
M 


dAbl CH/ 
TAR1-5-19CK 


HB2151 


Protein L 


0.1M glycine 
pH2.0 


Myc/flag 


RA~6n 
M 


TAR1-5-19CH/ 
dAb2 CK 


HB2151 


Protein L 


Total protein 


0.1M glycine pH2.0 


RA~8n 
M 

CA-12 
nM 


TAR1-5-19CH/ 
dAb3CK 


HB2151 


Protein L 


Total protein 


0.1M glycine pH2.0 


RA~3n 
M 



5 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

107 

PCR construction of TAR1-5-19CYS dimer 

See example 8 describing the dAb trimer. The trimer protocol gives rise to a mixture of 
monomer, dimer and trimer. 

5 Expression and purification of TAR1-5-19CYS dimer 

The dimer was purified from the supernatant of the culture by capture on Protein L 
agarose as outlined in the example 8. 

Separation of TAR1-5-19CYS monomer from the TAR1-5-19CYS dimer 

10 Prior to cation exchange separation, the mixed monomer/dimer sample was buffer 
exchanged into 50 mM sodium acetate buffer pH 4.0 using a PD-10 column (Amersham 
Pharmacia), following the manufacturer's guidelines. The sample was then applied to a 
lmL Resource S cation exchange column (Amersham Pharmacia), which had been pre- 
equilibrated with 50 mM sodium acetate pH 4.0. The monomer and dimer were separated 

15 using the following salt gradient in 50 mM sodium acetate pH 4.0: 

150 to 200 mM sodium chloride over 15 column volumes 
200 to 450 mM sodium chloride over 10 column volumes 
450 to 1000 mM sodium chloride over 15 column volumes 

20 

Fractions containing dimer only were identified using SDS-PAGE and then pooled and 
the pH increased to 8 by the addition of 1/5 volume of 1M Tris pH 8.0. 

In vitro functional binding assay: TNF receptor assay and cell assay 

25 The affinity of the dimer for human TNFce was determined using the TNF receptor and 
cell assay. IC50 in the receptor assay was approximately 0.3-0.8 nM; ND50 in the cell 
assay was approximately 3-8 nM. 

Other possible TAR1-5-19CYS dimer formats 

30 

PEG dimers and custom synthetic maleimide dimers 

Nektar (Shearwater) offer a range of bi-maleimide PEGs [mPEG2-(MAL)2 or mPEG- 
(MAL)2] which would allow the monomer to be formatted as a dimer, with a small linker 



WO 2004/003019 



PCT/GB2003/002804 



108 

separating the dAbs and both being linked to a PEG ranging in size from 5 to 40 kDa. It 
has been shown that the 5kDa mPEG-(MAL)2 (ie, [TARl-5-19]-Cys-maleimide-PEG x 
2, wherein the maleimides are linked together in the dimer) has an affinity in the TNF 
receptor assay of - 1-3 nM. Also the dimer can also be produced using TMEA (Tris[2- 
5 maleimidoethyl] amine) (Pierce Biotechnology) or other bi- functional linkers. 

It is also possible to produce the disulphide dimer using a chemical coupling procedure 
using 2 3 2'-dithiodipyridine (Sigma Aldrich) and the reduced monomer. 

10 Addition of a polypeptide linker or hinge to the C-terminus of the dAh. 

A small linker, either (Gly4Ser) n where n=l to 10, eg, 1, 2, 3, 4, 5, 6 or 7, an 
immunoglobulin (eg, IgG hinge region or random peptide sequence (eg, selected from a 
library of random peptide sequences) can be engineered between the dAb and the terminal 
cysteine residue. This can then be used to make dimers as outlined above. 

15 

Example 8 

dAb trimerisation 

20 

Summary 

For dAb trimerisation, a free cysteine is required at the C-terminus of the protein. The 
cysteine residue, once reduced to give the free thiol, can then be used to specifically 
25 couple the protein to a trimeric maleimide molecule, for example TMEA (Tris[2- 
maleimido ethyl] amine) . 

PGR construction of TAR1-5-19CYS 

The following oligonucleotides were used to specifically PGR TAR1-5-19 with a Sail and 
30 BamHL sites for cloning and also to introduce a C-terminal cysteine residue: 

Sail 

Trp Ser Ala Ser Thr Asp* He Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 
35 1 TGG AGC GCG TCG ACG GAC ATC CAG ATG ACC CAG TCT CCA TCC TCT CTG TCT GCA TCT GTA 

ACC TCG CGC AGC TGC CTG TAG GTC TAC TGG GTC AGA GGT AGG AGA GAC AGA CGT AGA CAT 



Gly Asp Arg Val Thr He Thr Cys Arg Ala Ser Gin Ser He Asp Ser Tyr Leu His Trp 



WO 2004/003019 



PCT/GB2003/002804 



109 

61 GGA GAC CGT GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC ATT GAT AGT TAT TTA CAT TGG 
CCT CTG GCA CAG TGG TAG TGA ACG GCC CGT TCA GTC TCG TAA CTA TCA ATA AAT GTA ACC 







Tyr 


Gin 


Gin 


Lys 


Pro 


Gly 


Lys 


Ala 


Pro 


Lys 


Leu 


Leu 


lie 


Tyr 


Ser 


Ala 


Ser 


Glu 


Leu 


Gin 


5 


121 


TAC 


CAG 


CAG 


AAA 


CCA 


GGG 


AAA 


GCC 


CCT 


AAG 


CTC 


CTG 


ATC 


TAT 


AGT 


GCA 


TCC 


GAG 


TTG 


CAA 






ATG 


GTC 


GTC 


TTT 


GGT 


CCC 


TTT 


CGG 


GGA 


TTC 


GAG 


GAC 


TAG 


ATA 


TCA 


CGT 


AGG 


CTC 




GTT 






Ser 


Gly 


Val 


Pro 


Ser 


Arg 


Phe 


Ser 


Gly 


Ser 


Gly 


Ser 


Gly 


Thr 


Asp 


Phe 


Thr 


Leu 


Thr 


lie 




181 


AGT 


GGG 


GTC 


CCA 


TCA 


CGT 


TTC 


AGT 


GGC 


AGT 


GGA 


TCT 


GGG 


ACA 


GAT 


TTC 


ACT 


CTC 


ACC 


ATC 


10 




TCA 


CCC 


CAG 


GGT 


AGT 


GCA 


AAG 


TCA 


CCG 


TCA 


CCT 


AGA 


CCC 


TGT 


CTA 


AAG 


TGA 


GAG 


TGG 


TAG 






Ser 


Ser 


Leu 


Gin 


Pro 


Glu 


Asp 


Phe 


Ala 


Thr 


Tyr 


Tyr 


Cys 


Gin 


Gin 


Val 


Val 


Trp 


Arg 


Pro 




241 


AGC 


AGT 


CTG 


CAA 


CCT 


GAA 


GAT 


TTT 


GCT 


ACG 


TAC 


TAC 


TGT 


CAA 


CAG 


GTT 


GTG 


TGG 


CGT 


CCT 






TCG 


TCA 


GAC 


GTT 


GGA 


CTT 


CTA 


AAA 


CGA 


TGC 


ATG 


ATG 


ACA 


GTT 


GTC 


CAA 


CAC 


ACC 


GCA 


GGA 


15 




































BamHI 










Phe 


Thr 


Phe 


Gly 


Gin 


Gly 


Thr 


Lys 


Val 


Glu 


lie 


Lys 


Arg 


Cys 


*** 


** * 


Gly 


Ser 


Gly 






301 


TTT 


ACG 


TTC 


GGC 


CAA 


GGG 


ACC 


AAG 


GTG 


GAA 


ATC 


AAA 


CGG 


TGC 


TAA 


TAA 


GGA 


TCC 


GGC 








AAA 


TGC 


AAG 


CCG 


GTT 


CCC 


TGG 


TTC 


CAC 


CTT 


TAG 


TTT 


GCC 


ACG 


ATT 


ATT 


CCT 


AGG 


CCG 





20 

(* start ofTARl-5-19CYS sequence) 



Forward primer 

25 5 ' -TGGAGCGCGTCGACGGAC ATCC AGATGACCC AGTCTCCA-3 ' 
Reverse primer 

5 ' -TTAGC AGCCGGATCCTTATT AGC ACCGTTTGATTTCC AC-3 5 

30 

The PGR reaction (50|liL volume) was set up as follows: 200|aM dNTPs, 0.4jliM of each 
primer, 5 [xL of lOx P/wTurbo buffer (Stratagene), 100 ng of template plasmid (encoding 
TAR1-5-19), l\xL of P/wTurbo enzyme (Stratagene) and the volume adjusted to 50jaL 
using sterile water. The following PCR conditions were used: initial denaturing step 94 
35 °C for 2 mins, then 25 cycles of 94 °C for 30 sees, 64 °C for 30 sec and 72 °C for 30 sec. 
A final extension step was also included of 72 °C for 5 mins. The PCR product was 
purified and digested with Sail and BamHI and ligated into the vector which had also 
been cut with the same restriction enzymes. Correct clones were verified by DNA 
sequencing. 

40 



WO 2004/003019 . PCT/GB2003/002804 

110 

Expression and purification of TAR1-5-19CYS 

TAR1-5-19CYS vector was transformed into BL21 (DE3) pLysS chemically competent 
cells (Novagen) following the manufacturer's protocol. Cells carrying the dAb plasmid 
were selected for using 100|Hg/mL carbenicillin and 37 \xg/mL chloramphenicol. Cultures 
5 were set up in 2L baffled flasks containing 500 mL of terrific broth (Sigma- Aldrich), 
100|ng/mL carbenicillin and 37 |xg/mL chloramphenicol. The cultures were grown at 30 
°C at 200rpm to an O.D.600 of 1-1.5 and then induced with ImM DPTG (isopropyl-beta- 
D-thiogalactopyranoside, from Melford Laboratories). The expression of the dAb was 
allowed to continue for 12-16 hrs at 30 °C. It was found that most of the dAb was present 

10 in the culture media. Therefore, the cells were separated from the media by centrifugation 
(8,000xg* for 30 mins), and the supernatant used to purify the dAb. Per litre of 
supernatant, 30 mL of Protein L agarose (Affitech) was added and the dAb allowed to 
batch bind with stirring for 2 hours. The resin was then allowed to settle under gravity for 
a further hour before the supernatant was siphoned off. The agarose was then packed into 

15 a XK 50 column (Amersham Phamacia) and was washed with 10 column volumes of 
PBS. The bound dAb was eluted with 100 mM glycine pH 2.0 and protein containing 
fractions were then neutralized by the addition of 1/5 volume of 1 M Tris pH 8.0. Per litre 
of culture supernatant 20 mg of pure protein was isolated, which contained a 50:50 ratio 
of monomer to dimer. 

20 

Trimerisation of TAR1-5-19CYS 

2.5 ml of 100 jjM TAR1-5-19CYS was reduce with 5 mM dithiothreitol and left at room 
temperature for 20 minutes. The sample was then buffer exchanged using a PD-10 
column (Amersham Pharmacia). The column had been pre-equilibrated with 5 mM 

25 EDTA, 50 mM sodium phosphate pH 6.5, and the sample applied and eluted following 

the manufactures guidelines. The sample was placed on ice until required. TMEA (Tris [2- 
maleimido ethyl] amine) was purchased from Pierce Biotechnology. A 20 mM stock 
solution of TMEA was made in 100% DMSO (dimethyl sulphoxide). It was found that a 
concentration of TMEA greater than 3:1 (molar ratio of dAb:TMEA) caused the rapid 

30 precipitation and cross-linking of the protein. Also the rate of precipitation and cross- 
linking was greater as the pH increased. Therefore using 100 pM reduced TAR1-5- 
19CYS, 25 |liM TMEA was added to trimerise the protein and the reaction allowed to 
proceed at room temperature for two hours. It was found that the addition of additives 



WO 2004/003019 



PCT/GB2003/002804 



111 

such as glycerol or ethylene glycol to 20% (v/v), significantly reduced the precipitation 
of the trimer as the coupling reaction proceeded. After coupling, SDS-PAGE analysis 
showed the presence of monomer, dimer and trimer in solution. 

5 Purification of the trimeric TAR1-5-19CYS 

40 jaL of 40% glacial acetic acid was added per mL of the TMEA-TAR1 - 5-1 9cys reaction 
to reduce the pH to -4. The sample was then applied to a lmL Resource S cation 
exchange column (Amersham Pharmacia), which had been pre-equilibrated with 50 niM 
sodium acetate pH 4.0. The dimer and trimer were partially separated using a salt gradient 
10 of 340 to 450 mM Sodium chloride, 50 mM sodium acetate pH 4.0 over 30 column 
volumes. Fractions containing trimer only were identified using SDS-PAGE and then 
pooled and the pH increased to 8 by the addition of 1/5 volume of 1M Tris pH 8.0. To 
prevent precipitation of the trimer during concentration steps (using 5K cut off Viva spin 
concentrators; Vivascience), 10% glycerol was added to the sample. 

15 

In vitro functional binding assay: TNF receptor assay and cell assay 
The affinity of the trimer for human TNFce was determined using the TNF receptor and 
cell assay. IC50 in the receptor assay was 0.3nM; ND50 in the cell assay was in the range 
of 3 to lOnM (eg, 3nM). 

20 

Other possible TAR1-5-19CYS trimer formats 

. TAR1-5-19CYS may also be formatted into a trimer using the following reagents: 

PEG trimers and custom synthetic maleimide trimers 

25 Nektar (Shearwater) offer a range of multi arm PEGs, which can be chemically modified 
at the terminal end of the PEG. Therefore using a PEG trimer with a maleimide functional 
group at the end of each arm would allow the trimerisation of the dAb in a manner similar 
to that outlined above using TMEA. The PEG may also have the advantage in increasing 
the solubility of the trimer thus preventing the problem of aggregation. Thus, one could 

30 produce a dAb trimer in which each dAb has a C-terminal cysteine that is linked to a 
maleimide functional group, the maleimide functional groups being linked to a PEG 
trimer. 



WO 2004/003019 PCT/GB2003/002804 

112 

Addition of a polypeptide linker or hinge to the C-terminus of the dAb 
A small linker, either (Gly 4 Ser) n where n= 1 to 10, eg, 1, 2, 3, 4, 5, 6 or 7 , an 
immunoglobulin (eg, IgG hinge region or random peptide sequence (eg, selected from a 
library of random peptide sequences) could be engineered between the dAb and the 
5 terminal cysteine residue. When used to make multimers (eg, dimers or trimers), this 
again would introduce a greater degree of flexibility and distance between the individual 
monomers, which may improve the binding characteristics to the target, eg a multisubunit 
target such as human TNFce. 

10 Example 9. 

Selection of a collection of single domain antibodies (dAbs) directed against human 
serum albumin (HSA) and mouse serum albumin (MSA). 

15 This example explains a method for making a single domain antibody (dAb) directed 
against serum albumin. Selection of dAbs against both mouse serum albumin (MSA) and 
human serum albumin (HSA) is described. Three human phage display antibody libraries 
were used in this experiment, each based on a single human framework for V H (see 
Figure 13: sequence of dummy V H based on V3-23/DP47 and JH4b) or Vk (see Figure 

20 15: sequence of dummy Vk based on ol2/o2/DPK9 and Jkl) with side chain diversity 
encoded by NNK codons incorporated in complementarity determining regions (CDR1 , 
CDR2 and CDR3). 

Library 1 (Vh): 

25 Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, 
H97, H98. 

Library size: 6.2 x 10 9 
Library 2 (Vh): 

30 Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, 
H97, H98, H99, H100, HlOOa, H100b. 
Library size: 4.3 x 10 9 



WO 2004/003019 



PCT/GB2003/002804 



113 

Library 3 (Vk): 

Diversity at positions: L30, L31, L32, L34, L50, L53, L91, L92, L93, L94, L96 
Library size: 2 x 10 9 

The Vh and Vk libraries have been preselected for binding to generic ligands protein A 
and protein L respectively so that the majority of clones in the unselected libraries are 
functional. The sizes of the libraries shown above correspond to the sizes after 
preselection. 

Two rounds of selection were performed on serum albumin using each of the libraries 
separately. For each selection, antigen was coated on immunotube (nunc) in 4ml of PBS 
at a concentration of 100/ig/ml. In the first round of selection, each of the three libraries 
was panned separately against HSA (Sigma) and MSA (Sigma). In the second round of 
selection, phage from each of the six first round selections was p aimed against (i) the 
same antigen again (eg 1 st round MSA, 2 nd round MSA) and (ii) against the reciprocal 
antigen (eg 1 st round MSA, 2 nd round HSA) resulting in a total of twelve 2 nd round 
selections. In each case, after the second round of selection 48 clones were tested for 
binding to HSA and MSA. Soluble dAb fragments were produced as described for scFv 
fragments by Harrison et al, Methods Enzymol. 1996;267:83-109 and standard ELISA 
protocol was followed (Hoogenboom et ah (1991) Nucleic Acids Res., 19: 4133) except 
that 2% tween PBS was used as a blocking buffer and bound dAbs were detected with 
either protein L-HRP (Sigma) (for the V/cs) and protein A -HRP (Amersham Pharmacia 
Biotech) (for the Vhs). 

dAbs that gave a signal above background indicating binding to MSA, HSA or both were 
tested in ELISA insoluble form for binding to plastic alone but all were specific for serum 
albumin. Clones were then sequenced (see table below) revealing that 21 unique dAb 
sequences had been identified. The minimum similarity (at the amino acid level) between 
the Vk dAb clones selected was 86.25% ((69/80)xl00; the result when all the diversified 
residues are different, eg clones 24 and 34). The minimum similarity between the V H dAb 
clones selected was 94 % ((127/136)xl00). 



WO 2004/003019 



PCT/GB2003/002804 



114 

Next, the serum albumin binding dAbs were tested for their ability to capture 
biotinylated antigen from solution. ELISA protocol (as above) was followed except that 
ELIS A plate was coated with 1 /ig/ml protein L (for the V/c clones) and 1 jug/ml protein A 
(for the Vh clones). Soluble dAb was captured from solution as in the protocol and 
5 detection was with biotinylated MSA or HSA and streptavidin HRP. The biotinylated 
MSA and HSA had been prepared according to the manufacturer's instructions, with the 
aim of achieving an average of 2 biotins per serum albumin molecule. Twenty four 
clones were identified that captured biotinylated MSA from solution in the ELISA. Two 
of these (clones 2 and 38 below) also captured biotinylated HSA. Next, the dAbs were 
10 tested for their ability to bind MSA coated on a CMS biacore chip. Eight clones were 
found that bound MSA on the biacore. 



dAb (all 

capture 

biotinylated 

MSA) 

V/c library 3 

template 

(dummy) 

2, 4, 7, 41, 

38, 54 

46, 47, 52, 56 

13,15 

30,35 

19, 
22, 
23, 
24, 

31, 
33, 

34, 
53, 

H> 
12, 
17, 
18, 

16, 21 
25, 26 
27, 
55, 

V H library 1 
(and 2) 
template 
(dummy) 
8,10 
36, 



H 

or k CDR1 



CDR2 



CDR3 



Binds 

MSA Captures 

in biotinylated 

biacore? HSA? 



K 


XXXLX 


XASXLQS 


QQXXXXPXT 




K 


SSYLN 


RASPLQS 


QQTYSVPPT 




K 


SSYLN 


RASPLQS 


QQTYRIPPT 




K 


FKSLK 


NASYLQS 


QQWYWPVT 




K 


YYHLK 


KASTLQS 


QQVRKVPRT 




K 


RRYLK 


QASVLQS 


QQGLYPPIT 




K 


YNWLK 


RASSLQS 


QQNWIPRT 




K 


LWHLR 


HASLLQS 


QQSAVYPKT 




K 


FRYLA 


HASHLQS 


QQRLLYPKT 




K 


FYHLA 


PASKLQS 


QQRARWPRT 




K 


IWHLN 


RASRLQS 


QQVARVPRT 




K 


YRYLR 


KASSLQS 


QQYVGYPRT 




K 


LKYLK 


NASHLQS 


QQTTYYPIT 




K 


LRYLR 


KASWLQS 


QQVLYYPQT 




K 


LRSLK 


AASRLQS 


QQWYWPAT 




K 


FRHLK 


AASRLQS 


QQVALYPKT 


V 


K 


RKYLR 


TASSLQS 


QQNLFWPRT 


S 


K 


RRYLN 


AASSLQS 


QQMLFYPKT 




K 


IKHLK 


GASRLQS 


QQGARWPQT 


s 


K 


YYHLK 


KASTLQS 


QQVRKVPRT 




K 


YKHLK 


NASHLQS 


QQVGRYPKT 




K 


FKSLK 


NASYLQS 


QQWYWPVT 




H 


XXYXXX 


XI XXXGXXTXYAD SVKG 


XXXX(XXXX) FDY 




H 


WVYQMD 


S I S AFGAKTLYADS VKG 


LSGKFDY 




H 


WSYQMT 


SISSFGSSTLYADSVKG 


GRDHNYSLFDY 





S all 4 bind 
S both bind 



WO 2004/003019 



PCT/GB2003/002804 



115 

In all cases the frameworks were identical to the frameworks in the corresponding 
dummy sequence, with diversity in the CDRs as indicated in the table above. 

Of the eight clones that bound MSA on the biacore, two clones that are highly expressed 
5 in E. coli (clones MSA16 and MSA26) were chosen for further study (see example 10). 
Full nucleotide and amino acid sequences for MSA16 and 26 are given in figure 16. 

Example 10. 

10 Determination of affinity and serum half-life in mouse of MSA binding dAbs MSA16 
and MSA26. 

dAbs MSA16 and MSA26 were expressed in the periplasm oiE. coli and purified using 
batch absorbtion to protein L-agarose affinity resin (Affitech, Norway) followed by 

15 elution with glycine at pH 2.2. The purified dAbs were then analysed by inhibition 
biacore to determine K d . Briefly, purified MSA16 and MSA26 were tested to determine 
the concentration of dAb required to achieve 200RUs of response on a biacore CMS chip 
coated with a high density of MSA. Once the required concentrations of dAb had been 
determined, MSA antigen at a range of concentrations around the expected Kd was 

20 premixed with the dAb and incubated overnight. Binding to the MSA coated biacore chip 
of dAb in each of the premixes was then measured at a high flow-rate of 30 jul/minute. 
The resulting curves were used to create Klotz plots, which gave an estimated Kd of 
200nM for MSA16 and 70nM for MSA 26 (Figure 17 A & B). 

25 Next, clones MSA16 and MSA26 were cloned into an expression vector with the HA tag 
(nucleic acid sequence: TATCCTTATGATGTTCCTGATTATGCA and amino acid 
sequence: YPYDVPDYA) and 2-10 mg quantities were expressed in E. coli and purified 
from the supernatant with protein L-agarose affinity resin (Affitech, Norway) and eluted 
with glycine at pH2.2. Serum half life of the dAbs was determined in mouse. MSA26 

30 and MSA16 were dosed as single i.v. injections at approx 1.5mg/kg into CD1 mice. 
Analysis of serum levels was by goat anti-HA (Abeam, UK) capture and protein L-HRP 
(invitrogen) detection ELISA which was blocked with 4% Marvel. Washing was with 
0.05% tween PBS. Standard curves of known concentrations of dAb were set up in the 



WO 2004/003019 



PCT/GB2003/002804 



116 

presence of lxmouse serum to ensure comparability with the test samples. Modelling 
with a 2 compartment model showed MSA-26 had a tl/2ce of 0.16hr ? a tl/2jS of 14.5hr and 
an area under the curve (AUC) of 465hr.mg/ml (data not shown) and MSA- 16 had a tl/2a 
of 0.98hr, a tl/2/3 of 36.5hr and an AUC of 913hr.mg/ml (figure 18). Both anti-MSA 
5 clones had considerably lengthened half life compared with HEL4 (an anti-hen egg white 
lysozymedAb) which had a tl/2a of 0.06hr> and a tl/2(3 of 0.34hr. 

Example 11. 

10 Creation of V h -Vh and Vk- Vk dual specific Fab like fragments 

This example describes a method for making V H ~ Vh and Vk-Vk dual specifics as Fab 
like fragments. Before constructing each of the Fab like fragments described, dAbs that 
bind to targets of choice were first selected from dAb libraries similar to those described 

15 in example 9. A V H dAb, HEL4, that binds to hen egg lysozyme (Sigma) was isolated 
and a second Vh dAb (TAR2h-5) that binds to TNFa receptor (R and D systems) was also 
isolated. The sequences of these are given in the sequence listing. A Vk dAb that binds 
TNFa; (TAR 1-5 -19) was isolated by selection and affinity maturation and the sequence is 
also set forth in the sequence listing. A second Vk dAb (MSA 26) described in example 9 

20 whose sequence is in figure 1 7B was also used in these experiments. 

DNA from expression vectors containing the four dAbs described above was digested 
with enzymes Sail and NotI to excise the DNA coding for the dAb. A band of the 
expected size (300-400bp) was purified by running the digest on an agarose gel and 
25 excising the band, followed by gel purification using the Qiagen gel purification kit 
(Qiagen, UK). The DNA coding for the dAbs was then inserted into either the Ch or Ck 
vectors (Figs 8 and 9) as indicated in the table below. 



dAb 


Target antigen 


dAb V H or 
dAb Vk 


Inserted into 
vector 


tag(C 
terminal) 


Antibiotic 
resisitance 


HEL4 


Hen egg lysozyme 


V h 


Ch 


myc 


Chloramphenicol 


TAR2-5 


TNF receptor 


V h 


Ck 


flag 


Ampicillin 


TAR1-S-19 


TNF a 


Vk 


C h 


myc 


Chloramphenicol 


MSA 26 


Mouse serum albumin 


Vk 


Ck 


flag 


Ampicillin 



WO 2004/003019 PCT/GB2003/002804 

117 

The V H C h and V H Ck constructs were cotransformed into HB2151 cells. Separately, the 
V/c C H and Vk Ck constructs were cotransformed into HB2151 cells. Cultures of each of 
the cotransformed cell lines were grown overnight (in 2xTy containing 5% glucose, 
10/xg/ml chloramphenicol and 100/xg/ml ampicillin to maintain antibiotic selection for 
5 both C H and Ck plasmids). The overnight cultures were used to inoculate fresh media 
(2xTy, 10/xg/ml chloramphenicol and 100/xg/ml ampicillin) and grown to OD 0.7-0.9 
before induction by the addition of IPTG to express their C H and Ck constructs. 
Expressed Fab like fragment was then purified from the periplasm by protein A 
purification (for the contransformed V H C H and V H Ck) and MSA affinity resin 
10 purification (for the contransformed Vk Ch and Vk Ck). 

Vh-Vh dual specific 

Expression of the V H Ch and V H Ck dual specific was tested by running the protein on a 
gel. The gel was blotted and a band the expected size for the Fab fragment could be 

15 detected on the Western blot via both the myc tag and the flag tag, indicating that both the 
V H C h and V H Ck parts of the Fab like fragment were present. Next, in order to 
determine whether the two halves of the dual specific were present in the same Fab-like 
fragment, an ELISA plate was coated overnight at 4°C with 100 /xl per well of hen egg 
lysozyme (HEL) at 3 mg/ml in sodium bicarbonate buffer. The plate was then blocked 

20 (as described in example 1) with 2% tween PBS followed by incubation with the V H C H 
/V H Ck dual specific Fab like fragment. Detection of binding of the dual specific to the 
HEL was via the non cognate chain using 9el0 (a monoclonal antibody that binds the 
myc tag, Roche) and anti mouse IgG-HRP (Amersham Pharmacia Biotech). The signal 
for the V H C h /V h Ck dual specific Fab like fragment was 0.154 compared to a 

25 background signal of 0.069 for the V H Ck chain expressed alone. This demonstrates that 
the Fab like fragment has binding specificity for target antigen. 

V K -V K dual specific 

After purifying the contransformed Vk C h and Vk Ck dual specific Fab like fragment on 
30 an MSA affinity resin, the resulting protein was used to probe an ELISA plate coated with 
1/xg/ml TNFa and an ELISA plate coated with 10/xg/ml MSA. As predicted, there was 
signal above background when detected with protein L-HRP on bot ELISA plates (data 
not shown). This indicated that the fraction of protein able to bind to MSA (and therefore 



WO 2004/003019 PCT/GB2003/002804 

118 

purified on the MSA affinity column) was also able to bind TNFa in a subsequent 
ELISA, confirming the dual specificity of the antibody fragment. This fraction of protein 
was then used for two subsequent experiments. Firstly, an ELISA plate coated with 
, 1/xg/ml TNFa was probed with dual specific Vtc C H and Vk Ck Fab like fragment and 
5 also with a control TNFa binding dAb at a concentration calculated to give a similar 
signal on the ELISA. Both the dual specific and control dAb were used to probe the 
ELISA plate in the presence and in the absence of 2mg/ml MSA. The signal in the dual 
specific well was reduced by more than 50% but the signal in the dAb well was not 
reduced at all (see figure 19a). The same protein was also put into the receptor assay with 
10 and without MSA and competition by MSA was also shown (see figure 19c). This 
demonstrates that binding of MSA to the dual specific is competitive with binding to 
TNFo;. 



15 Example 12. 

Creation of a Vk- Vic dual specific cys bonded dual specific with specificity for 
mouse serum albumin and TNFa 

20 This example describes a method for making a dual specific antibody fragment specific 
for both mouse serum albumin and TNFo; by chemical coupling via a disulphide bond. 
Both MSA16 (from example 1) and TAR1-5-19 dAbs were recloned into a pET based 
vector with a C terminal cysteine and no tags. The two dAbs were expressed at 4-10 mg 
levels and purified from the supernatant using protein L-agarose affinity resin (Affitiech, 

25 Norway). The cysteine tagged dAbs were then reduced with dithiothreitol. The TAR1-5- 
1 9 dAb was then coupled with dithiodipyridine to block reformation of disulphide bonds 
resulting in the formation of PEP 1-5-19 homodimers. The two different dAbs were then 
mixed at pH 6.5 to promote disulphide bond formation and the generation of TAR1-5-19, 
MSA16 cys bonded heterodimers. This method for producing conjugates of two unlike 

30 proteins was originally described by Bang et al (King TP, Li Y Kochoumian L 
Biochemistry. 1978 voll7: 1499-506 Preparation of protein conjugates via intermolecular 
disulfide bond formation.) Heterodimers were separated from monomelic species by 
cation exchange. Separation was confirmed by the presence of a band of the expected 



WO 2004/003019 PCT/GB2003/002804 

119 

size on a SDS gel. The resulting heterodimeric species was tested in the TNF receptor 
assay and found to have an IC50 for neutralising TNF of approximately 18 nM. Next, 
the receptor assay was repeated with a constant concentration of heterodimer (18nM) and 
a dilution series of MSA and HSA. The presence of HSA at a range of concentrations (up 
5 to 2 mg/ml) did not cause a reduction in the ability of the dimer to inhibit TNFo? . 
However, the addition of MSA caused a dose dependant reduction in the ability of the 
dimer to inhibit TNFo: (figure 20).This demonstrates that MSA and TNFa compete for 
binding to the cys bonded TAR1-5-19, MSA16 dimer. 

10 Data Summary 

A summary of data obtained in the experiments set forth in the foregoing examples is set 
forth in Annex 4. 

All publications mentioned in the present specification, and references cited in said 
15 publications, are herein incorporated by reference. Various modifications and variations 
of the described methods and system of the invention will be apparent to those skilled in 
the art without departing from the scope and spirit of the invention. Although the 
invention has been described in connection with specific preferred embodiments, it 
should be understood that the invention as claimed should not be unduly limited to such 
20 specific embodiments. Indeed, various modifications of the described modes for carrying 
out the invention which are obvious to those skilled in molecular biology or related fields 
are intended to be within the scope of the following claims. 



WO 2004/003019 



PCT/GB2003/002804 



120 

Annex 1; polypeptides which enhance half-life in vivo. 

Alpha- 1 Glycoprotein (Orosomucoid) (AAG) 

Alpha- 1 Antichyromotrypsin (ACT) 

Alpha- 1 Antitrypsin (AAT) 

Alpha- 1 Microglobulin (Protein HC) (AIM) 

Alpha-2 Macroglobulin (A2M) 

Antithrornbin III (AT III) 

Apolipoprotein A-l (Apo A-l) 

Apoliprotein B (Apo B) 

Beta-2-microglobulin (B2M) 

Ceruloplasmin (Cp) 

Complement Component (C3) 

Complement Component (C4) 

CI Esterase Inhibitor (CI INH) 

C-Reactive Protein (CRP) 

Cystatin C (Cys C) 

Ferritin (FER) 

Fibrinogen (FIB) 

Fibronectin (FN) 

Haptoglobin (Hp) 

Hemopexin (HPX) 

Immunoglobulin A (IgA) 

Immunoglobulin D (IgD) 

Immunoglobulin E (IgE) 

Immunoglobulin G (IgG) 

Immunoglobulin M (IgM) 

Immunoglobulin Light Chains (kapa/lambda) 

Lipoprotein(a) [Lp(a)] 

Mannose-bindign protein (MBP) 

Myoglobin (Myo) 

Plasminogen (PSM) 

Prealbumin (Transthyretin) (PAL) 



WO 2004/003019 PCT/GB2003/002804 

121 

Retinol-binding protein (RBP) 
Rheomatoid Factor (RF) 
Serum Amyloid A (SAA) 
Soluble Tranferrin Receptor (sTfR) 
5 Transferrin (Tf) 



WO 2004/003019 



PCT/GB2003/002804 



122 

Annex 2 



Pairing 


Therapeutic relevant references. 


TNF 

ALPHA/TGF-p 


• TGF-b and TNF when injected into the ankle joint of collagen induced 
arthritis model significantly enhanced joint inflammation. In non-collagen 
challenged mice there was no effect. 


TNF ALPHA/IL- 
1 


• TNF and IL-1 synergize in the pathology of uveitis. 

• TNF and IL-1 synergize in the pathology of malaria (hypoglycaemia, NO). 

• TNF and IL-1 synergize in the induction of polymorphonuclear (PMN) 
cells migration in inflammation. 

• IL-1 and TNF synergize to induce PMN infiltration into the peritoneum. 

• IL-1 and TNF synergize to induce the secretion of IL-1 by endothelial cells. 
Important in inflammation. 

• IL-1 or TNF alone induced some cellular infiltration into knee synovium. 
IL-1 induced PMNs, TNF - monocytes. Together they induced a more 
severe infiltration due to increased PMNs. 

• Circulating myocardial depressant substance (present in sepsis) is low 
levels of IL-1 andTNFacting synergistically. 


TNF ALPHA/IL-2 


• Most relating to synergistic activation of killer T-cells. 


TNF ALPHA/IL-3 


• Synergy of interleukin 3 and tumor necrosis factor alpha in stimulating 
clonal growth of acute myelogenous leukemia blasts is the result of 
induction of secondary hematopoietic cytokines by tumor necrosis factor 
alpha. 

• Cancer Res. 1992 Apr 15;52(8):2 197-201. 


TNF ALPHA/IL-4 


• IL-4 and TNF svnergize to induce VCAM expression on endothelial cells. 
Implied to have a role in asthma. Same for synovium - implicated in RA. 

• TNF and IL-4 synergize to induce IL-6 expression in keratinocytes. 

• Sustained elevated levels of VCAM- 1 in cultured fibroblast-like 
synoviocytes can be achieved by TNF-alpha in combination with either IL- 
4 or IL-1 3 through increased mRNA stability. Am J Pathol. 1999 
Apr;154(4):l 149-58 


TNF ALPHA/IL-5 


• Relationship between the tumor necrosis factor system and the serum 

mterleukin-4, interleukin-5 9 interleukin- 8, eosinophil cationic protein, and 
immunoglobulin E levels in the bronchial hyperreactivity of adults and 
their children. Allergy Asthma Proc. 2003 Mar-Apr;24(2): 111-8. 


TNF ALPHA/IL-6 


• TNF and IL-6 are potent growth factors for OH-2, a novel human myeloma 

ST C 

cell line. Ear J Haematol 1994 Jul;53(l):3 1-7. 


TNF ALPHA/IL-8 


• TNF and IL-8 synergized with PMNs to activate platelets. Implicated in 
Acute Respiratory Distress Syndrome. 

• See IL-5/TNF (asthma). Synergism between interleukin-8 and tumor 
necrosis factor-alpha for neutrophil-mediated platelet activation. Eur 
Cytokine Netw. 1994 Sep-Oct;5(5):455-60. (adult respiratory distress 
syndrome (ARDS)) 


TNF ALPHA/IL-9 




TNF ALPHA/IL- 
10 


• IL-10 induces and synergizes with TNF in the induction of HIV expression 
in chronically infected T-cells. 


TNF ALPHA/IL- 
11 


• Cytokines synergistically induce osteoclast differentiation: support by 
immortalized or normal calvarial cells. Am J Physiol Cell Physiol 2002 
Sep;283(3):C679-87. (Bone loss) 


TNF ALPHA/IL- 
12 





WO 2004/003019 



PCT/GB2003/002804 



123 



TNF ALPHA/IL- 
13 


• Sustained elevated levels of VCAM-1 in cultured fibroblast-like 
synoviocytes can be achieved by TNF-alpha in combination with either IL- 
4 or IL-13 through increased rnRNA stability. Am J Pathol. 1999 
Apr;154(4):l 149-58. 

• Interleukin-13 and tumour necrosis factor-alpha synergistically induce 
eotaxin production in human nasal fibroblasts. Clin Exp Allergy. 2000 
Mar;30(3):348-55. 

• Interleukin-13 and tumour necrosis factor-alpha synergistically induce 
eotaxin production in human nasal fibroblasts. Clin Exp Allergy. 2000 
Mar;30(3):348-55 (allergic inflammation) 

• Implications of serum TNF -beta and IL-13 in the treatment response of 
childhood nephrotic syndrome. Cytokine. 2003 Feb 7;21(3):155-9. 


TNF ALPHA/IL- 
14 


• Effects of inhaled tumour necrosis factor alpha in subjects with mild 
asthma. Thorax. 2002 Sep;57(9):774-8. 


TNF ALPHA/IL- 
15 


• Effects of inhaled tumour necrosis factor alpha in subjects with mild 
asthma. Thorax. 2002 Sep;57(9):774-8. 


TNF ALPHA/IL- 
16 


• Tumor necrosis factor-alpha-induced synthesis of interleukin-16 in airway 
epithelial cells: priming for serotonin stimulation. Am J Respir Cell Mo I 
Biol. 2003 Mar;28(3):354-62. (airway inflammation) 

• Correlation of circulating interleukin 1 6 with proinflammatory cytokines in 
patients with rheumatoid arthritis. Rheumatology (Oxford). 2001 
Apr;40(4):474-5. No abstract available. 

• Interleukin 1 6 is up-regulated in Crohn's disease and participates in TNBS 
colitis in mice. Gastroenterology. 2000 Oct;119(4):972-82. 


TNF ALPHA/IL- 
17 


• Inhibition of interleukin- 17 prevents the development of arthritis in 
vaccinated mice challenged with Borrelia burgdorferi. Infect Immun. 2003 
Jun;71(6):3437-42. 

• Interleukin 17 synergises with tumour necrosis factor alpha to induce 
cartilage destruction in vitro. Ann Rheum Dis. 2002 Oct;61(10):870-6. 

• A role of GM-CSF in the accumulation of neutrophils in the airways caused 
byIL-17 and TNF-alpha. Eur Respir J. 2003 Mar;21(3):387-93. (Airway 
inflammation) 

• Abstract Interleukin- 1, tumor necrosis factor alpha, and interleukin- 17 
synergistically up-reeulate nitric oxide and prostaglandin E2 production in 
explants of human osteoarthritic knee menisci. Arthritis Rheum. 2001 
Sep;44(9):2078-83. 


TNF ALPHA/IL- 
18 


• Association of interleukin- 18 expression with enhanced levels of both 
interleukin- lbeta and tumor necrosis factor alpha in knee synovial tissue of 
patients with rheumatoid arthritis. Arthritis Rheum. 2003 Feb;48(2):339- 
47. 

• Abstract Elevated levels of interleukin- 1 8 and tumor necrosis factor-alpha 
in serum of patients with type 2 diabetes mellitus: relationship with diabetic 
nephropathy. Metabolism. 2003 May;52(5): 605-8. 


TNF ALPHA/IL- 
19 


• Abstract IL-19 induces production of IL-6 and TNF-alpha and results in 
cell apoptosis through TNF-alpha. J Immunol. 2002 Oct 15;169(8):4288- 
97. 


TNF ALPHA/IL- 
20 


• Abstract Cytokines: IL-20 - a new effector in skin inflammation. Curr Biol. 
2001 Jull0;ll(13):R531-4 


TNF 

ALPHA/Complem 
ent 


• Inflammation and coagulation: implications for the septic patient. Clin 
Infect Dis. 2003 May 15;36(10): 1259-65. Epub 2003 May 08. Review. 


TNF 

ALPHA/IFN-y 


• MHC induction in the brain. 

• Synergize in anti- viral response/IFN-p induction. 

• Neutrophil activation/ respiratory burst. 

• Endothelial cell activation 

• Toxicities noted when patients treated with TNF/IFN-y as anti-viral therapy 



WO 2004/003019 



PCT/GB2003/002804 



124 





• Fractalkine expression by human astrocytes. 




• Many papers on inflammatory responses - i.e. LPS, also macrophage 




activation. 




• Anti-TNF and anti-IFN-y synergize to protect mice from lethal 




endotoxemia. 



TGF-p/IL-1 


• Prostaglndin synthesis by osteoblasts 

• IL-6 production by intestinal epithelial cells (inflammation model) 

• Stimulates IL-1 1 and IL-6 in lung fibroblasts (inflammation model) 

• IL-6 and IL-8 production in the retina 


TGF-p/IL-6 


• Chondrocarcoma proliferation 


IL-l/IL-2 


• B-cell activation 

• LAK cell activation 

• T-cell activation 

• IL-1 synergy with IL-2 in the generation of lymphokine activated killer 
cells is mediated by TNF-alpha and beta (lymphotoxin). Cytokine. 1992 
Nov;4(6):479-87. 


IL-l/IL-3 




IL-l/IL-4 


• B-cell activation 

• IL-4 induces IL-1 expression in endothelial cell activation. 


IL-l/IL-5 




IL-l/IL-6 


• B cell activation 

• T cell activation (can replace accessory cells) 

• IL-1 induces IL-6 expression 

JL 

• C3 and serum amyloid expression (acute phase response) 

• HIV expression 

• Cartilage collagen breakdown. 


IL-l/IL-7 


• IL-7 is requisite for IL-1 -induced thymocyte proliferation. Involvement of 
IL-7 in the synergistic effects of granulocyte-macrophage colony- 
stimulating factor or tumor necrosis factor with IL-1. J Immunol. 1992 Jan 
1;148(1):99-105. 


IL-l/IL-8 




IL-l/IL-10 




TT _1/TT _1 1 


• (~VtnlHnp<! <?vnerffi stic all v induce osteoclast differentiation: sutTDortbv 
immortalized or normal calvarial cells. Am J Physiol Cell Physiol 2002 
Sep;283(3):C679-87. (Bone loss) 


IL-l/IL-16 


• Correlation of circulating interleukin 1 6 with proinflammatorv cytokines in 
patients with rheumatoid arthritis. Rheumatology (Oxford). 2001 
Apr;40(4):474-5. No abstract available. 


IL-l/IL-17 


• Inhibition of interleukin- 17 prevents the development of arthritis in 
vaccinated mice challenged with Borrelia burgdorferi. Infect Immun. 2003 
Jun;71(6):3437-42. 

• Contribution of interleukin 17 to human cartilage degradation and synovial 
inflammation in osteoarthritis. Osteoarthritis Cartilage. 2002 




Oct;10(10):799-807. 
• Abstract Interleukin- 1, tumor necrosis factor alpha, and interleukin- 17 

synergistically up-regulate nitric oxide and prostaglandin E2 production in 
explants of human osteoarthritic knee menisci. Arthritis Rheum. 2001 
Sep;44(9):2078-83. 


IL-l/IL-18 


• Association of interleukin- 1 8 expression with enhanced levels of both 

interleukin- lbeta and tumor necrosis factor alpha in knee synovial tissue of 
patients with rheumatoid arthritis. Arthritis Rheum. 2003 Feb;48(2):339-47. 


IL-l/IFN-g 




IL-2/IL-3 


• T-cell proliferation 

• B cell proliferation 



WO 2004/003019 



PCT/GB2003/002804 



125 



IL-2/IL-4 


• B~cell proliferation 

• T-cell proliferation 

• (selectively inducing activation of CD8 and NK lymphocytes)IL-2R beta 
agonist Pl-30 acts in synergy with IL-2, IL-4, IL-9, and IL-15: biological 
and molecular effects. J Immunol 2000 Oct 15;165(8): 4312-8. 


IL-2/IL-5 


• B-cell proliferation/ Ig secretion 

• IL-5 induces IL-2 receptors on B-cells 


IL-2/IL-6 


• Development of cytotoxic T-cells 


IL-2/IL-7 




IL-2/IL-9 


• See IL-2/IL-4 (NK-cells) 


IL-2/IL-10 


• B-cell activation 


IL-2/IL-12 


• IL-12 synergizes with IL-2 to induce lymphokine- activated cytotoxicity 
and perforin and granzyme gene expression in fresh human NK cells. Cell 
Immunol 1995 Oct l;165(l):33-43. (T-cell activation) 


IL-2/IL-15 


• See IL-2/IL-4 (NK cells) 

• (T cell activation and proliferation) IL-15 and IL-2: a matter of life and 
death for T cells in vivo. Nat Med. 2001 Jan;7(l): 114-8. 


IL-2/IL-16 


• Synergistic activation of CD4+ T cells by IL-16 and IL-2. J Immunol 1998 
Mar 1;160(5):2115-20. 


IL-2/IL-17 


• Evidence for the early involvement of interleukin 17 in human and 

experimental renal allograft rejection. J Pathol 2002 Jul; 197(3): 3 22-3 2. 


IL-2/IL-18 


• Interleukin 18 (IL-18) in synergy with IL-2 induces lethal lung injury in 
mice: a potential role for cytokines, chemokines, and natural killer cells in 
the pathogenesis of interstitial pneumonia. Blood. 2002 Feb 15;99(4):1289- 
98. 


IL-2/TGF-p 


• Control of CD4 effector fate: transforming growth factor beta 1 and 
interleukin 2 synergize to prevent apoptosis and promote effector 
expansion. J Exp Med. 1995 Sep l;182(3):699-709. 


IL-2/IFN-y 


• Ig secretion by B-cells 

• IL-2 induces IFN-y expression by T-cells 


IL-2/IFN-cc/|3 


• None 


IL-3/IL-4 


• Synergize in mast cell growth 

• Synergistic effects of IL-4 and either GM-CSF or IL-3 on the induction of 
CD23 expression by human monocytes: regulatory effects of IFN-alpha and 
IFN-gamma. Cytokine. 1994 Jul;6(4):407-13. 


IL-3/IL-5 




IL-3/IL-6 




TT -3/IFN-v 


• IL-4 and IFN-gamma synergistically increase total polymeric IgA receptor 
levels in human intestinal epithelial cells. Role of protein tyrosine kinases. 
J Immunol 1996 Jun 15;156(12):4807-14. 


IL-3/GM-CSF 


• Differential regulation of human eosinophil IL-3, IL-5, and GM-CSF 
receptor alpha-chain expression by cytokines: IL-3, IL-5, and GM-CSF 
down-regulate IL-5 receptor alpha expression with loss of IL-5 
responsiveness, but up-regulate IL-3 receptor alpha expression. J Immunol 
2003 Jun l;170(ll):5359-66. (allergic inflammation) 


IL-4/IL-2 


• IL-4 synergistically enhances both IL-2- and IL-12-induced IFN- {gamma} 
expression in murine NK cells. Blood. 2003 Mar 13 [Epub ahead of print] 


IL-4/IL-5 


• Enhanced mast cell histamine etc. secretion in response to IgE 

• A Th2-like cytokine response is involved in bullous pemphigoid, the role of 
IL-4 and IL-5 in the pathogenesis of the disease. Int J Immunopathol 
Pharmacol. 1999 May-Aug;12(2):55-61. 


IL-4/IL-6 




IL-4/IL-10 




IL-4/IL-11 


• Synergistic interactions between interleukin- 1 1 and interleukin-4 in support 
of proliferation of primitive hematopoietic progenitors of mice. Blood. 



WO 2004/003019 



PCT/GB2003/002804 



126 





1991 Sepl5;78(6):1448-51. 


IL-4/IL-12 


• Synergistic effects of IL-4 and IL- 1 8 on IL- 1 2-dependent IFN-garnma 
production by dendritic cells. J Immunol 2000 Jan 1;164(1):64-71. 
(increase Thl/Th2 differentiation) 

• IL-4 synergistically enhances both IL-2- and IL-12-induced IFN- {gamma} 
expression in murine NK cells. Blood. 2003 Mar 13 [Epub ahead of print] 


IL-4/IL-13 


• Abstract Interleukin-4 and interleukin-13 signaling connections maps. 
Science. 2003 Jun 6;300(5625): 1527-8. (allergy, asthma) 

• Inhibition of the IL-4/IL-13 receptor system prevents allergic sensitization 
without affecting established allergy in a mouse model for allergic asthma. 
J Allergy Clin Immunol. 2003 Jun;lll(6): 1361-1369. 


IL-4/IL-16 


• (asthma) Interleukin (IL)-4/IL-9 and exogenous IL-16 induce IL-16 
production by BEAS-2B cells, a bronchial epithelial cell line. Cell 
Immunol. 2001 Feb l;207(2):75-80 


IL-4/IL-17 


• Interleukin (IL)-4 and IL-17 synergistically stimulate IL-6 secretion in 
human colonic myofibroblasts, hit J Mol Med. 2002 Nov;10(5):631-4. 
(Gut inflammation) 


IL-4/IL-24 


• IL-24 is expressed by rat and human macrophages. Immunobiology. 2002 
Jul;205(3):321-34. 


IL-4/IL-25 


• Abstract New IL-17 family members promote Thl or Th2 responses in the 
lung: in vivo function of the novel cytokine IL-25. J Immunol. 2002 Jul 
l;169(l):443-53. (allergic inflammation) 

• Abstract Mast cells produce interleukin-25 upon Fcepsilon Rl-mediated 
activation. Blood. 2003 May l;101(9):3594-6. Epub 2003 Jan 02. (allergic 
inflammation) 


IL-4/IFN-Y 


• Abstract Interleukin 4 induces interleukin 6 production by endothelial cells: 
synergy with interferon-gamma. Eur J Immunol. 1991 Jan;21 ( 1 ) : 97- 1 0 1 . 


IL-4/SCF 


• Regulation of human intestinal mast cells by stem cell factor and IL-4. 
Immunol Rev. 2001 Feb; 179: 5 7-60. Review. 


IL-5/IL-3 


• Differential regulation of human eosinophil IL-3, IL-5, and GM-CSF 
receptor alpha-chain expression by cytokines: IL-3, IL-5, and GM-CSF 
down-regulate IL-5 receptor alpha expression with loss of IL-5 
responsiveness, but up-regulate IL-3 receptor alpha expression. J Immunol. 
2003 Jun 1;170(1 1):5359-66. (Allergic inflammation see abstract) 


IL-5/IL-6 




IL-5/IL-13 


• Inhibition of allergic airways inflammation and airway 

hyperresponsiveness in mice by dexamethasone: role of eosinophils, IL-5, 
eotaxin, andIL-13. J Allergy Clin Immunol. 2003 May;ll 1(5): 1049-61. 


IL-5/IL-17 


• Interleukin- 17 orchestrates the granulocyte influx into airways after 

allergen inhalation in a mouse model of allergic asthma. Am J Respir Cell 
Mol Biol. 2003 Jan;28(l):42-50. 


IL-5/IL-25 


• Abstract New IL-17 family members promote Thl or Th2 responses in the 
lung: in vivo function of the novel cytokine IL-25. J Immunol. 2002 Jul 
l;169(l):443-53. (allergic inflammation) 

• Abstract Mast cells produce interleukin-25 upon Fcepsilon Rl-mediated 
activation. Blood. 2003 May l;101(9):3594-6. Epub 2003 Jan 02. (allergic 
inflammation) 


IL-5/IFN-y 




IL-5/GM-CSF 


• Differential regulation of human eosinophil IL-3, IL-5, and GM-CSF 
receptor alpha-chain expression by cytokines: IL-3, IL-5, and GM-CSF 
down-regulate IL-5 receptor alpha expression with loss of IL-5 
responsiveness, but up-regulate IL-3 receptor alpha expression. J Immunol. 
2003 Jun l;170(ll):5359-66. (Allergic inflammation) 


IL-6/IL-10 




IL-6/IL-11 




IL-6/IL-16 


• Interleukin- 16 stimulates the expression and production of pro- 



WO 2004/003019 



PCT/GB2003/002804 



127 





inflammatory cytokines by human monocytes. Immunology. 2000 
May;100(l):63-9. 


IL-6/IL-17 


• Stimulation of airway mucin gene expression by interleukin (IL)-17 
through IL-6 paracrine/autocrine loop. J Biol Chem. 2003 May 
9;278(19): 17036-43. Epub 2003 Mar 06. (airway inflammation, asthma) 


IL-6/IL-19 


• Abstract IL-19 induces production of IL-6 and TNF-alpha and results in 
cell apoptosis through TNF-alpha. J Immunol 2002 Oct 15;169(8):4288- 
97. 


IL-6/IFN-g 




IL-7/IL-2 


• Interleukin 7 worsens graft- versus-host disease. Blood. 2002 Oct 
l;100(7):2642-9. 


IL-7/IL-12 


• Synergistic effects of IL-7 and IL-12 on human T cell activation. J 
Immunol 1995 May 15;154(10):5093-102. 


IL-7/IL-15 


• Interleukin-7 and interleukin- 15 regulate the expression of the bcl-2 and c- 
myb genes in cutaneous T-cell lymphoma cells. Blood. 2001 Nov 
l;98(9):2778-83. (growth factor) 


IL-8/IL-11 


• Abnormal production of interleukin (IL)-1 1 and IL-8 in polycythaemia 
vera. Cytokine. 2002 Nov 2 1;20(4): 178-83. 


IL-8/IL-17 


• The Role of IL-17 in Joint Destruction. Drug News Per sped. 2002 
Jan;15(l): 17-23. (arthritis) 

• Abstract Interleukin- 17 stimulates the expression of interleukin- 8, growth- 
related oncogene-alpha, and granulocyte-colony-stimulating factor by 
human airway epithelial cells. Am J Respir Cell Mo I Biol 2002 
Jun;26(6):748-53. (airway inflammation) 


IL-8/GSF 


• Interleukin-8: an autocrine/paracrine growth factor for human 

hematopoietic progenitors acting in synergy with colony stimulating factor- 
1 to promote monocyte-macrophage growth and differentiation. Exp 
Hematol. 1999 Jan;27(l):28-36. 


IL-8/VGEF 


• Intracavitary VEGF, bFGF, IL-8, IL-12 levels in primary and recurrent 
malignant glioma. JNeurooncol 2003 May;62(3):297-303. 


IL-9/IL-4 


• Anti-interleukin-9 antibody treatment inhibits airway inflammation and 
hyperreactivity in mouse asthma model. Am J Respir Crit Care Med. 2002 
Aug 1;166(3):409-16. 


IL-9/IL-5 


• Pulmonary overexpression of IL-9 induces Th2 cytokine expression, 
leading to immune pathology. J Clin Invest. 2002 Jan;109(l):29-39. 

• Th2 cytokines and asthma. Interleukin-9 as a therapeutic target for asthma. 
Respir Res. 2001;2(2):80-4. Epub 2001 Feb 15. Review. 

• Abstract Interleukin-9 enhances interleukin-5 receptor expression, 
differentiation, and survival of human eosinophils. Blood. 2000 Sep 
15:96(61:2163-71 (asthma) 


IL-9/IL-13 


• Anti-interleukin-9 antibody treatment inhibits airway inflammation and 
hyperreactivity in mouse asthma model. Am J Respir Crit Care Med. 2002 
Aug 1;166(3):409-16. 

• Direct effects of interleukin- 13 on epithelial cells cause airway 
hyperreactivity and mucus overproduction in asthma. Nat Med. 2002 
Aug;8(8):885-9. 


IL-9/IL-16 


• See IL-4/IL-16 


IL-10/IL-2 


• The interplay of interleukin- 1 0 (IL-1 0) and interleukm-2 (IL-2) in humoral 
immune responses: IL-10 synergizes with IL-2 to enhance responses of 
human B lymphocytes in a mechanism which is different from upregulation 
of CD25 expression. Cell Immunol. 1994 Sep;157(2):478-88. 


IL-10/IL-12 




IL-10/TGF-P 


• IL-10 and TGF-beta cooperate in the regulatory T cell response to mucosal 
allergens in normal immunity and specific immunotherapy. Eur J 
Immunol. 2003 May;33(5): 1205-14. 


IL-10/IFN-y 





WO 2004/003019 



PCT/GB2003/002804 



128 



IL-ll/IL-6 


• Interleukin-6 and interleukin-1 1 support human osteoclast formation by a 
RANKL-independent mechanism. Bone, 2003 Jan;32(l):l-7. (bone 
resorption in inflammation) 


IL-ll/IL-17 


• Polarized in vivo expression of IL-1 1 and IL-17 between acute and chronic 
skin lesions. J Allergy Clin Immunol 2003 Apr;lll(4):875-81. (allergic 
dermatitis) 

• IL-17 promotes bone erosion in murine collagen-induced arthritis through 
loss of the receptor activator of NF-kappa B ligand/osteoprotegerin 
balance. J Immunol. 2003 Mar l;170(5):2655-62. 


IL-11/TGF-P 


• Polarized in vivo expression of IL-1 1 and IL-17 between acute and chronic 
skin lesions. J Allergy Clin Immunol 2003 Apr;lll(4):875-81. (allergic 
dermatitis) 


IL-12/IL-13 


• Relationship of Interleukin-1 2 and Interleukin-1 3 imbalance with class- 
specific rheumatoid factors and anticardiolipin antibodies in systemic lupus 
erythematosus. Clin Rheumatol 2003 May;22(2):107-ll. 


IL-12/IL-17 


• Upregulation of interleukin- 1 2 and - 1 7 in active inflammatory bowel 
disease. Scand J Gastroenterol 2003 Feb;38(2): 180-5. 


IL-12/IL-18 


• Synergistic proliferation and activation of natural killer cells by interleukin 
12 and interleukin 18. Cytokine. 1999 Nov;ll(l l):822-30. 

• Inflammatory Liver Steatosis Caused by IL-12 and IL-18. J Interferon 
Cytokine Res. 2003 Mar;23(3): 155-62. 


IL-12/IL-23 


• nterleukin-23 rather than interleukin- 12 is the critical cytokine for 
autoimmune inflammation of the brain. Nature. 2003 Feb 
13;421(6924):744-8. 

• Abstract A unique role for IL-23 in promoting cellular immunity. J Leukoc 
Biol 2003 Jan;73(l):49-56. Review. 


IL-12/IL-27 


• Abstract IL-27, a heterodimeric cytokine composed of EBB and p28 
protein, induces proliferation of naive CD4(+) T cells. Immunity. 2002 
Jun;16(6):779-90. 


IL-12/IFN-y 


• IL-12 induces IFN-y expression by B and T-cells as part of immune 
stimulation. 


IL-13/IL-5 


• See IL-5/IL-13 


IL-13/IL-25 


• Abstract New IL-17 family members promote Thl or Th2 responses in the 
lung: in vivo function of the novel cytokine IL-25. J Immunol 2002 Jul 
l;169(l):443-53. (allergic inflammation) 

• Abstract Mast cells produce interleukin-25 upon Fcepsilon Rl-mediated 
activation. Blood. 2003 May l;101(9):3594-6. Epub 2003 Jan 02. (allergic 
inflammation) 


IL-15/IL-13 


• Differential expression of interleukins (IL)-13 and IL-15 in ectopic and 
eutopic endometrium of women with endometriosis and normal fertile 
women. Am J Reprod Immunol 2003 Feb;49(2):75-83. 


IL-15/IL-16 


• IL-15 and IL-1 6 overexpression in cutaneous T-cell lymphomas: stage- 
dependent increase in mycosis fungoides progression. Exp Dermatol 2000 
Aug;9(4):248-51. 


IL-15/IL-17 


• Abstract IL-17, produced by lymphocytes and neutrophils, is necessary for 
lipopolysaccharide-induced airway neutrophilia: IL-15 as a possible trigger. 
J Immunol. 2003 Feb 15;170(4):2106-12. (airway inflammation) 


IL-15/IL-21 


• IL-2 1 m Synergy with IL-1 5 or IL-1 o Lnnances lr JN -gamma rroauction m 
Human NK and T Cells. J Immunol 2003 Jun 1;170(1 1):5464-9. 


IL-17/IL-23 


• Interleukin-23 promotes a distinct CD4 T cell activation state characterized 
by the production of interleukin- 17. J Biol Chem. 2003 Jan 
17;278(3): 1910-4. Epub 2002 Nov 03 


IL-17/TGF-P 


• Polarized in vivo expression of IL-1 1 and IL-17 between acute and chronic 
skin lesions. J Allergy Clin Immunol 2003 Apr;lll(4):875-81. (allergic 
dermatitis) 


IL-18/IL-12 


• Svnergistic proliferation and activation of natural killer cells by interleukin 



WO 2004/003019 



PCT/GB2003/002804 



129 





12 and interleukin 18. Cytokine. 1999 Nov;ll(l l):822-30. 
• Abstract Inhibition of in vitro immunoglobulin production by IL-12 in 
murine chronic graft-vs.-host disease: synergism with IL-18. Eur J 
Immunol. \yyo Jun ? Zo(oj.zui /-z*t. 


IL-18/IL-21 


• IL-2 1 in Synergy with IL-1 5 or IL-1 8 Enhances IFN-gamma Production in 
Human NK and T Cells. J Immunol. 2003 Jun l;170(ll):5464-9. 


IL-18/TGF-P 


• Interleukin 18 and transforming growth factor betal in the serum of 
patients with Graves' ophthalmopathy treated with corticosteroids. Int 
Immunopharmacol. 2003 Apr;3(4): 549-52. 


IL-18/IFN-Y 




Anti-TNF 
ALPHA/anti-CD4 


• Synergistic therapeutic effect in DBA/1 arthritic mice. 



WO 2004/003019 

Annex 3: Oncology combinations 



130 



PCT/GB2003/002804 



Target 


Disease 


Pair with 


CD89* 


Use as cytotoxic cell recruiter 


all 








CD19 


B cell lymphomas 


HLA-DR 






CD5 


HLA-DR 


B cell lymphomas 


CD89 






CD19 






CD5 


CD38 


Multiple myeloma 


CD138 






CDS 6 






HLA-DR 


CD138 


Multiple myeloma 


CD38 






CD56 






HLA-DR 


CD138 


Lung cancer 


CD56 






CEA 


CD33 


Acute myelod lymphoma 


CD34 






HLA-DR 


CD56 


Lung cancer 


CD138 






CEA 


CEA 


Pan carcinoma 


MET receptor 


VEGF 


Pan carcinoma 


MET receptor 


VEGF 


Pan carcinoma 


MET receptor 


receptor 






IL-13 


Asthma/pulmonary 


IL-4 




inflammation 


IL-5 






Eotaxin(s) 






MDC 






TARC 






TNFoc 






IL-9 






EGFR 






CD40L 






IL-25 






MCP-1 






TGFP 


EL-4 


Asthma 


IL-13 






IL-5 






Eotaxin(s) 






MDC 






TARC 






TNFa 






IL-9 






EGFR 






CD40L 






IL-25 






MCP-1 






TGFp 


Eotaxin 


Asthma 


IL-5 






Eotaxin-2 






Eotaxin-3 


EGFR 


cancer 


HER2/neu 






HER3 






HER4 


HER2 


cancer 


HER3 



WO 2004/003019 



PCT/GB2003/002804 



131 







HER4 


TNFR1 


RA/Crohn's disease 


IL-IR 
IL-6R 
IL-18R 




A/OroTvn's disease 


TT -loc/6 

t..» ' J. UArf I-J 

IL-6 

IL-18 

ICAM-1 

IL-15 

IL-17 


IL-1R 


RA/Crohn's disease 


IL-6R 
IL-18R 


IL-18R 


RA/Crohn's disease 


IL-6R 



WO 2004/003019 



132 



PCT/GB2003/002804 



co 

s 

co 

ca 



Q co 
C co 



o 
in 

O 



o 
o 

in 



a 



CO 
< 



i 
g 



CO 

< 



O 



T 



wo 
i — i 

II 



CN O 

i — I r— l 
II II 



> — i 
II 



OS 

Q 



en 
wo 



ITl 

u 



o 



i 

o 

1=1 

I 



o 
o 
m 



CO 



s 



H 

CO 





<N oo M 
_ H I! H 

^(^ On ^ 

T^-? I I I 

rh in in in 
i i i 



^ H H H 



oo 
II 

ON 

i — i 
i 

in 
i 



ii u 

o o 

1 — I *— I 

U O 

ON On 



OO CO 
II II 



o o 

cs co 

o o 

on on 



i 



i 



in m «n in 



in 



^ H ^ H 



3 



O 

© 



o 



in 



6 

a 
I 



CO 



O 



H 

CO 



C o 

■a a 

eo 7^ 

s II 

s § 

"3 ^ 



| 

in Pi o 
o o - 

l| cn 2 S 
m w in in 



s 

o 

a 

o 



CO 

< 



H 

CO 



O 
CO 



t— I 

2 



52 

a 
§ 



co 
*© 



wo 
i 



ON 
1 

in 
i 



a 

o 

el 

o 

B 




On 



co 

B 



o 

B 
Id 



H 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



133 



o 
vo 

II 



CD 



« o o o p 

m co «o m co 

go II !) U I! 

*3 ^3 $3 

O»o uo w~> to 

. I I I 1 

f-H f— I §— I f— I 



o o 
o o 

II II 

to vo 
to *o 



in 



o 
o 

1— 1 

II 

O 

S3 

*j 
o 

in 
i 



toJQ 

03 



o 



Q 



O 

o 

lO 



o 



o 
o 
io 



=5. 
1 — I 

p 



^ o 

o 1—1 
o ^ 
io 2 

O *cd 

<4-( 







s 

o 






"a 

o 
o 

CO 



< 

CO 
I 

I 



CO 

I 

O 

I 



I 

< 

oo 



00 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



134 

Claims 

1 . A dual-specific ligand comprising a first immunoglobulin single variable 
domain having a binding specificity to a first epitope or antigen and a second 
complementary immunoglobulin single variable domain having a binding activity 
to a second epitope or antigen, wherein one or both of said antigens or epitopes 
acts to increase the half-life of the ligand in vivo and wherein said first and second 
domains lack mutually complementary domains which share the same specificity, 
provided that said dual specific ligand does not consist of an anti-HS A V H domain 
and an anti-p galactosidase V K domain. 

2. A dual-specific ligand according to claim 1, comprising at least one single 
heavy chain variable domain of an antibody and one complementary single light 
chain variable domain of an antibody such that the two regions are capable of 
associating to form a complementary VH/VL pair. 

3 . A dual specific ligand according to claim 2 wherein the V H and V L are 
provided by an antibody scFv fragment. 

4. A dual-specific ligand according to claim 2 wherein the V H and V L are 
provided by an antibody Fab region. 

5. A four chain IgG immunoglobulin ligand comprising a dual specific 
ligand of claim 2. 

6. A four chain IgG immunoglobulin ligand according to claim 5, wherein 
said IgG comprises two dual specific ligands, said dual specific ligands being 
identical in their variable domains. 

7. A four chain IgG immunoglobulin ligand according to claim 5, wherein 
said IgG comprises two dual specific ligands, said dual specific ligands being 
different in their variable domains. 

8. A ligand comprising a first immunoglobulin variable domain having a first 
antigen or epitope binding specificity and a second immunoglobulin variable 
domain having a second antigen or epitope binding specificity wherein one or 
both of said first and second variable domains bind to an antigen which increases 
the half-life of the ligand in vivo, and the variable domains are not complementary 
to one another. 

9. A ligand according to claim 8 wherein the first and the second 
immunoglobulin variable domains are heavy chain variable domains (V H ). 

10. A ligand according to claim 8 wherein the first and the second 
immunoglobulin variable domains are light chain variable domains (V L ). 

11. A ligand according to claim any preceding claim, wherein the first and 
second epitopes bind independently, such that the dual specific ligand may 
simultaneously bind both the first and second epitopes or antigens. 



SU^fif iSWifi^^UlL^ 26) 



WO 2004/003019 



135 



PCT/GB2003/002804 



12. A ligand according to claim 11, wherein the dual specific ligand comprises 
a first form and a second form in equilibrium in solution, wherein both epitopes or 
antigens bind to the first form independently but compete for binding to the 
second form. 

13. A ligand according to any preceding claim wherein the variable regions 
are derived from immunoglobulins directed against said epitopes or antigens. 

14. A ligand according to any preceding claim, wherein said first and second 
epitopes are present on separate antigens. 

15. A ligand according to any one of claims 1 to 1 1, wherein said first and 
second epitopes are present on the same antigen. 

16. A ligand according to any preceding claim comprising a variable domain 
that is derived from a repertoire of single antibody domains. 

17. A ligand of claim 16 wherein said repertoire is displayed on the surface of 
filamentous bacteriophage and wherein the single antibody domains are selected 
by binding of the bacteriophage repertoire to antigen. 

18. A ligand according to any preceding claim wherein the sequence of at 
least one variable domain is modified by mutation or DNA shuffling. 

19. A dual-specific ligand according to any preceding claim wherein the 
variable regions are non-covalently associated. 

20. A dual-specific ligand according to any one of claims 1 to 1 8 wherein the 
variable regions are covalently associated. 

21 . A dual-specific ligand according to claim 20 wherein the covalent 
association is mediated by disulphide bonds. 

22. A dAb monomer ligand specific for TNFa, which dissociates from human 
TNFa with a dissociation constant (K d ) of 50nM to 20pM, and a K Q ff rate constant 
of 5xl0 _1 to lxlO" 7 s" 1 , as determined by surface plasmon resonance. 

23. A dAb monomer ligand specific for TNFa according to claim 22, wherein 
the dAb is a Vk. 

24. A dAb monomer ligand specific for TNF receptor 1 (p55), which 
dissociates from human TNF receptor 1 with a dissociation constant (K d ) of 50nM 
to 20pM, and aKoff rate constant of 5x1 0" 1 to lxl 0" 7 s" 1 , as determined by surface 
plasmon resonance. 

25. A dAb monomer ligand according to claim 22 to claim 24, wherein the 
monomer neutralises human TNFa or TNF receptor 1 in a standard cell assay 
with anND50 of 500nM to 50pM. 



WO 2004/003019 



136 



PCT/GB2003/002804 



26. A dAb monomer ligand specific for TNF receptor 1 (p55), wherein the 
dAb antagonises the activity of the TNF receptor 1 in a standard cell assay with 
an ND 50 of <100nM, and at a concentration of <10pM the dAb agonises the 
activity of the TNF receptor 1 by <5% in the assay. 

27. A dAb monomer ligand specific for serum albumin (SA) which dissociates 
from SA with a dissociation constant (K d ) of InM to SOO^M, as determined by 
surface plasmon resonance. 

28. A dAb monomer ligand according to claim 27, wherein the monomer 
binds SA in a standard ligand binding assay with an IC50 of InM to 500jaM. 

29. A dAb monomer ligand specific for TNFoc, wherein the dAb comprises 
the amino acid sequence of TAR1-5-19 or a sequence that is at least 80% 
homologous thereto. 

30. A dAb monomer ligand specific for TNFoc, wherein the dAb comprises 
the amino acid sequence of TAR1-5 or a sequence that is at least 80% 
homologous thereto. 

31. A dAb monomer ligand specific for TNFa, wherein the dAb comprises 
the amino acid sequence of TAR1-27 or a sequence that is at least 80% 
homologous thereto. 

32. A dAb monomer ligand specific for TNF receptor 1, wherein the dAb 
comprises the amino acid sequence of TAR2-10 or a sequence that is at least 80% 
homologous thereto. 

33. A dAb monomer ligand specific for TNF receptor 1, wherein the dAb 
comprises the amino acid sequence of TAR2-10 or a sequence that is at least 90% 
homologous thereto. 

34. A dAb monomer ligand specific for TNF receptor 1 , wherein the dAb 
comprises the amino acid sequence of TAR2-5 or a sequence that is at least 80% 
homologous thereto. 

35. A dAb monomer ligand specific for TNF receptor 1 , wherein the dAb 
comprises the amino acid sequence of TAR2-5 or a sequence that is at least 90% 
homologous thereto. 

36. A dAb monomer ligand specific for SA, wherein the dAb comprises the 
amino acid sequence of MSA- 16 or a sequence that is at least 80% homologous 
thereto. 

37. A dAb monomer ligand specific for SA, wherein the dAb comprises the 
amino acid sequence of MSA-26 or a sequence that is at least 80% homologous 
thereto. 



WO 2004/003019 



PCT/GB2003/002804 



137 

38. A dAb monomer according to any one of claims 29 to 37, wherein the 
TNFa, TNF receptor 1 or S A is in human form. 

39. A dAb monomer further comprising a terminal Cys residue. 

40. A dAb monomer according to any one of claims 29 to 38 5 further 
comprising a terminal Cys residue. 

41. A dual specific ligand comprising at least one dAb monomer according to 
any one of claims 22 to 40. 

42. A dual specific ligand according to claim 41, which is a dimer. 

43 . A dual specific ligand according to claim 42, wherein the dimer comprises 
anti-human TNF alpha dAb according to any one of claims 22, 23 and 28-30, and 
an anti-SA dAb according to any one of claims 26, 27 and 34-36. 

44. A dual specific ligand according to claim 42, wherein the dimer is a 
homo- or hetero-dimer comprising first and second anti-human TNF alpha dAbs, 
each dAb being according to any one of claims 22, 23 and 28-30. 

45. A dual specific ligand according to claim 41, which is a trimer. 

46. A dual specific ligand according to claim 45, which is a homotrimer 
comprising three copies of an anti-human TNF alpha dAb according to any one of 
claims 22, 23 and 29-3 1 . 

47. A ligand according to any preceding claim, which comprises a universal 
framework. 

48. A ligand according to claim 47, wherein the universal framework 
comprises a Vh framework selected from the group consisting of DP47, DP45 and 
DPS 8; and/or the V L framework is DPK9. 

49. A ligand according to any preceding claim which comprises a binding site 
for a generic ligand. 

50. The ligand of claim 49, wherein the generic ligand binding site is selected 
from the group consisting of protein A, protein L and protein G. 

51. A ligand according to any preceding claim, wherein the ligand comprises a 
variable domain having one or more framework regions comprising an amino acid 
sequence that is the same as the amino acid sequence of a corresponding 
framework region encoded by a human germline antibody gene segment, or the 
amino acid sequences of one or more of said framework regions collectively 
comprises up to 5 amino acid differences relative to the amino acid sequence of 
said corresponding framework region encoded by a human germline antibody 
gene segment. 



WO 2004/003019 



PCT/GB2003/002804 



138 

52. A ligand according to any one of claims 1 to 51 5 wherein the ligand 
comprises a variable domain, wherein the amino acid sequences of FW1, FW2, 
FW3 and FW4 are the same as the amino acid sequences of corresponding 
framework regions encoded by a human germline antibody gene segment, or the 
amino acid sequences of FW1, FW2, FW3 and FW4 collectively contain up to 10 
amino acid differences relative to the amino acid sequences of corresponding 
framework regions encoded by said human germline antibody gene segment. 

53. The ligand according to claim 51 or claim 52, which comprises an 
antibody variable domain comprising FW1, FW2 and FW3 regions, and the 
amino acid sequence of said FW1, FW2 and FW3 are the same as the amino acid 
sequences of corresponding framework regions encoded by human germline 
antibody gene segments. 

54. The ligand according to any one of claims 51 to 53, wherein said human 
germline antibody gene segment is selected from the group consisting of DP47, 
DP45, DP48 and DPK9. 

55. A ligand according to any preceding claim, comprising a V H domain that 
is not a Camelid immunoglobulin variable domain. 

56. The ligand of Claim 55, comprising a V H domain that does not contain one 
or more amino acids that are specific to Camelid immunoglobulin variable 
domains as compared to human Vh domains. 

57. A method for producing a ligand comprising a first immunoglobulin single 
variable domain having a first binding specificity and a second single 
immunoglobulin single variable domain having a second binding specificity, one 
or both of the binding specificities being specific for a protein which increases the 
half-life of the ligand in vivo, the method comprising the steps of: 

(a) selecting a first variable domain by its ability to bind to a first epitope, 

(b) selecting a second variable region by its ability to bind to a second 
epitope, 

(c) combining the variable regions; and 

(d) selecting the ligand by its ability to bind to said first and second epitopes; 
wherein, when said variable domains are complementary, neither of said domains 
is a Vh domain specific for HSA. 

58. A method according to claim 57 wherein said first variable domain is 
selected for binding to said first epitope in absence of a complementary variable 
domain. 

59. A method according to claim 57 wherein said first variable domain is 
selected for binding to said first epitope in the presence of a third complementary 
variable domain in which said third variable domain is different from said second 
variable domain. 

60. Nucleic acid encoding a dual-specific ligand according to any one of 
claims 1 to 56. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

139 

61 . A nucleic acid according to claim 60 which is specific for TNFoc, 
comprising the nucleic acid sequence of TAR1-5-19 or a sequence that is at least 
70% homologous thereto. 

62. A nucleic acid according to claim 60 which is specific for TNFa, 
comprising the nucleic acid sequence of TAR1-5 or a sequence that is at least 
70% homologous thereto. 

63 . A nucleic acid according to claim 60 which is specific for TNFa, 
comprising the nucleic acid sequence of TAR 1-27 or a sequence that is at least 
70% homologous thereto. 

64. A nucleic acid according to claim 60 which is specific for TNF receptor 1, 
comprising the nucleic acid sequence of TAR2-10 or a sequence that is at least 
70% homologous thereto. 

65. A nucleic acid according to claim 60 which is specific for TNF receptor 1 5 
comprising the nucleic acid sequence of TAR2-10 or a sequence that is at least 
80% homologous thereto. 

66. A nucleic acid according to claim 60 which is specific for TNF receptor 1 , 
comprising the nucleic acid sequence of TAR2h-5 or a sequence that is at least 
70% homologous thereto. 

67. A nucleic acid according to claim 60 which is specific for TNF receptor 1, 
comprising the nucleic acid sequence of TAR2h-5 or a sequence that is at least 
80% homologous thereto. 

68. A nucleic acid according to claim 60 which is specific for SA, comprising 
the nucleic acid sequence of MSA- 16 or a sequence that is at least 70% 
homologous thereto. 

69. A nucleic acid according to claim 60 which is specific for SA, comprising 
the nucleic acid sequence of MSA-26 or a sequence that is at least 70% 
homologous thereto. 

70. A vector comprising nucleic acid according to any one of claims 60 to 69. 

71 . A vector according to claim 70, further comprising components necessary 
for the expression of a dual-specific ligand. 

72. A host cell transfected with a vector according to claim 71 . 

73 . A method for producing a closed conformation multi-specific ligand 
comprising a first single epitope binding domain having a first epitope binding 
specificity and a non-complementary second epitope binding domain having a 
second epitope binding specificity, wherein the first and second binding 
specificities are capable of competing for epitope binding such that the closed 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



140 

conformation multi-specific ligand may not bind both epitopes simultaneously, 
said method comprising the steps of: 

a) selecting a first epitope binding domain by its ability to bind to a first 
epitope, 

b) selecting a second epitope binding domain by its ability to bind to a 
second epitope, 

c) combining the epitope binding domains such that the domains are in a 
closed conformation; and 

d) selecting the closed conformation multispecific ligand by its ability to bind 
to said first second epitope and said second epitope, but not to both said first and 
second epitopes simultaneously. 

74. A method according to claim 73 wherein the first and the second epitope 
binding domains are immunoglobulin variable heavy chain domains (Vh)* 

75. A method according to claim 73 wherein the first and the second 
immunoglobulin variable domains are immunoglobulin variable light chain 
domains (Vl)- 

76. A method according to any one of claims 73 to 75 wherein the 
immunoglobulin domains are derived from immunoglobulins directed against said 
epitopes. 

77. A method according to any one of claims 73 to 76, wherein said first and 
second epitopes are present on separate antigens. 

78. A method according to any one of claims 73 to 76, wherein said first and 
second epitopes are present on the same antigen. 

79. A method according to any one of claims 73 to 78 wherein the variable 
domain is derived from a repertoire of single antibody domains. 

80. A method of claim 79 wherein said repertoire is displayed on the surface 
of filamentous bacteriophage and wherein the single antibody domains are 
selected by binding of the bacteriophage repertoire to antigen. 

81 . A method of any one of claims 73 to 80 wherein the sequence of at least 
one immunoglobulin variable domain is modified by mutation or DNA shuffling. 

82. A closed conformation multispecific ligand comprising a first epitope 
binding domain having a first epitope binding specificity and a non- 
complementary second epitope binding domain having a second epitope binding 
specificity wherein the first and second binding specificities are capable of 
competing for epitope binding such that the closed conformation multi-specific 
ligand cannot bind both epitopes simultaneously. 

83. A closed conformation multispecific ligand according to claim 82, 
obtainable by a method according to any one of claims 73 to 80. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



141 



PCT/GB2003/002804 



84. A closed conformation multispecific ligand according to claim 82 or claim 
83, comprising more than one single heavy chain variable domain of an antibody 
or more than one light chain variable domain of an antibody. 

85. A closed conformation multi-specific ligand according to claim 84 
wherein the Vh an d Vl are linked by a peptide linker. 

86. A closed conformation multi-specific ligand according to claim 84 
wherein the Vh or Vl are provided by an antibody Fab-like region. 

87. A closed conformation multi-specific ligand according to any one of 
claims 82 to 84 wherein the variable regions are non-covalently associated. 

88. A closed conformation multi-specific ligand according to any one of 
claims 82 to 84 wherein the variable regions are covalently associated. 

89. A closed conformation multi-specific ligand according to claim 87 
wherein the covalent association is mediated by disulphide bonds. 

90. A closed conformation multi-specific ligand according to any of claims 82 
to 89 which comprises a universal framework. 

91. A closed conformation multi-specific ligand according to any of claims 82 
to 90 which comprises a binding site for a generic ligand. 

92. The closed conformation multi-specific ligand of claim 91 , wherein the 
generic ligand binding site is selected from the group consisting of protein A, 
protein L and protein G. 

93. A closed conformation ligand according to any of claims 82 to 92, 
wherein the ligand comprises a variable domain having one or more framework 
regions comprising an amino acid sequence that is the same as the amino acid 
sequence of a corresponding framework region encoded by a human germline 
antibody gene segment, or the amino acid sequences of one or more of said 
framework regions collectively comprises up to 5 amino acid differences relative 
to the amino acid sequence of said corresponding framework region encoded by a 
human germline antibody gene segment. 

94. The closed conformation ligand according to claim 93, wherein the ligand 
comprises a variable domain wherein the amino acid sequences of FW1, FW2, 
FW3 and FW4 are the same as the amino acid sequences of corresponding 
framework regions encoded by a human germline antibody gene segment, or the 
amino acid sequences of FW1, FW2, FW3 and FW4 collectively contain up to 10 
amino acid differences relative to the amino acid sequences of corresponding 
framework regions encoded by said human germline antibody gene segment. 

95. The closed conformation ligand according to claim 93 or claim 94, which 
comprises an antibody variable domain comprising FW1, FW2 and FW3 regions, 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



142 

and the amino acid sequence of said FW1, FW2 and FW3 are the same as the 
amino acid sequences of corresponding framework regions encoded by human 
germline antibody gene segments. 

96. The closed conformation ligand according to any one of claims 92 to 95, 
wherein said human germline antibody gene segment is selected from the group 
consisting of DP47, DP45, DP48 and DPK9. 

97. A closed conformation ligand according to any one of claims 92 to 96, 
comprising a Vh domain that is not a Camelid immunoglobulin variable domain. 

98. The closed conformation ligand of Claim 97 ? wherein the V H domain does 
not contain one or more amino acids that are specific to Camelid immunoglobulin 
variable domains as compared to human Vh domains. 

99. A closed conformation multi-specific ligand according to any one of 
claims 82 to 98, wherein one specificity thereof is for an agent effective to 
increase the half life of the ligand. 

100. A kit comprising a closed conformation multi-specific ligand according to 
any one of claims 82 to 99. 

101. Nucleic acid encoding at least a closed conformation multispecific ligand 
according to any one of claims 82 to 99. 

1 02. A vector comprising nucleic acid according to claim 101. 

103. A vector according to claim 102, further comprising components 
necessary for the expression of a closed conformation multispecific ligand. 

104. A host cell transfected with a vector according to claim 103. 

105. A method for detecting the presence of a target molecule, comprising: 

(a) providing a closed conformation multispecific ligand bound to an agent, said 
ligand being specific for the target molecule and the agent, wherein the agent 
which is bound by the ligand leads to the generation of a detectable signal on 
displacement from the ligand; 

(b) exposing the closed conformation multispecific ligand to the target molecule; 
and 

(c) detecting the signal generated as a result of the displacement of the agent. 

106. A method according to claim 105, wherein the agent is an enzyme, which 
is inactive when bound by the closed conformation multispecific ligand. 

107. A method according to claim 105, wherein the agent is the substrate for an 
enzyme 

108. A method according to claim 107, wherein the agent is a fluorescent, 
luminescent or chromogenic molecule which is inactive or quenched when bound 
by the ligand. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



143 



PCT/GB2003/002804 



109. A kit for performing a method according to any one of claims 105-108, 
comprising a closed conformation multispecific ligand capable of binding to a 
target molecule, and optionally an agent and buffers suitable therefor. 

110. A homogenous immunoassay incorporating a method according to any 
one of claims 105-108. 

111. A ligand according to any one of claims 1 to 56 for use in therapy. 

112. A pharmaceutical composition comprising a ligand according to any one 
of claims 1 to 56 ? and a pharmaceutical^ acceptable eccipient, carrier or diluent. 

113. A method for preparing a chelating multimeric ligand comprising the steps 
of: 

(a) providing a vector comprising a nucleic acid sequence encoding a 
single binding domain specific for a first epitope on a target; 

(b) providing a vector encoding a repertoire comprising second binding 
domains specific for a second epitope on said target, which epitope can be the 
same or different to the first epitope, said second epitope being adjacent to said 

first epitope; and 

(c) expressing said first and second binding domains; and 

(d) isolating those combinations of first and second binding domains which 
combine together to produce a target-binding dimer. 

114. A method according to claim 113, wherein the first and second binding 
domains are associated covalently through a linker. 

115. A method according to claim 113, wherein the first and second binding 
domains are associated non-covalently. 

116. A method according to claim 113, wherein the first and second binding 
domains are associated through natural association of the domains. 

117. A method according to claim 116, wherein the binding domains comprise 
a Vh domain and a V K domain. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



1 / 17 



o CJ 
PO W (5 
ffi < 

Eh 
Uj EH 
H 



CJ 
H 
Eh 



hi 



CD 



< 

o cd 
cd 

H 
CO CJ 
Eh 

O 
< CJ 

cd 

< 
cd 

EH 

o cd 

Eh 

CJ 
CO o 
Eh 

o O 
CM t-q EH 
ffi CJ 

Cd CD 

< 

CD 
i— I EH 
CJ 

CJ 
CO CJ 
Eh 

CD 
CD CD 
CD 



o 



ffi 



CJ 
Eh 



CJ 



Eh CJ 



CO 



CD 



CJ 
CD 
< 

EH 
CD 
IL2J 



CD CD 
CD 



CO 



cd CD 



CM 

ID CO 
ffi 



Eh 

CD 

Eh 
CD 

e3 

CD 



EH 
M EH 



o 

in r=C 

ffi 



o 

CD_ 



cm 

Cd 

a 

ffi 



o 



< 

CO o 
EH 

CJ 
> EH 
CD 

CD 
IS CD 
Eh 

CD 
CJ pH 
CD 



rH 
ffi 



CJ 

a < 

CD 
CD 
CD 
CJ 

< o 

CD 

<< 
Cd CD 
< 

CD 

H-l EH 

O 



CJ 

XI CO CD 



CM CD 
oo S H 
ffi < 



CJ 



o 
oo 

ffi 



CD 
i— I EH 

CJ 

Eh 

EH 

CD 
v—l Eh 
CJ 

CD 
Eh CJ 
< 

CJ 



ffi 





























CD 












o 




CD 


o 




CD 


CD 








CD 




Eh 




H 






CD 




CD 












<; 




CJ 


J— 1 


CJ 


CD 


CJ 








CJ 




CD 




Eh 








CD 






Q 




— | 


Eh 


CO 


CD 






< 




CD 








CD 




o 




< 








CD 














o 




o 






O 


CD 






CD 


CD 




r* . 

H 


CO 


CD 




1— 1 


Eh 




CD 








CD 




a 










< 


T hi 
CD 


b-H 








< 


yU 


CD 




CJ 








CD 






> 


Eh 




1 — ! 


Eh 










CD 








CD 














CD 




CJ 








EH 




Eh 




CJ 






CD 


CO 


CD 






CO 


r \ 

O 


CM 


O 


Eh 


CJ 




> 


H 




< 












o 










CD 






M 
























CD 


O 










CJ 










CD 


CO 




— i 






CJ 


CJ 


— i 


Eh 




< 




Eh 


X 






CD 




CD 




CJ 






< 




CJ 








Eh 


kd 






Eh 






CJ 


> 


H 






CO 


CJ 




EH 


CJ 




EH 


a 




CD 








EH 














< 




CJ 








CD 




CD 




o 






CD 


EH 


O 






1—1 


Eh 


CD 


CD 




Eh 




CD 


CD 




< 








CJ 




CD 




Eh 






CD 






















O 








CJ 




o 




CJ 


o 


<! 


o 


Eh 


O 




< 


> 


H 




t — 1 


CO 


CJ 




CJ 




< 


i— 1 


O < 




CD 








Eh 




CJ 




CD 






CJ 




CD 








CJ 














O 




Eh 






CO 


CJ 




EH 


CJ 




CD 


CD 












tH 








err 1 






CD 




O 












CD 




CD 






O 


EH 


CJ 






Ot 


CJ 






CD 


CD 






Eh 


< 








o 




CJ 




CD 






Eh 




< 








EH 




CD 




Eh 






CD 


CD 


CD 






CO 


CJ 


o 


< 


CO 


CJ 




Eh 


CJ 




CD 








EH 




a 




Eh 






< 




CD 








CD 




Eh 




< 








a 


< 






o» 


< 






CD 


CD 










U 








CJ 




EH 




CD 










u 








CJ 




CD 




EH 






Eh 


CD 


CD 






Eh 


o 


IS 


CD 


CO 


CD 




Cm 


CJ 




CD 












Eh 










CJ 



CD 
CD 
EH 





CD 


CD 






CJ 






a 




CD 


CD 


l-H" EH 




CO 


CJ 






< 






CD 


a 






EH 






Eh 






EH 


CD 






Eh 






O 




Cm 


CJ 


CD CD 




S3 


< 




Q 


< 






CJ 


CD 












CD 
















O 










CD 


CD 








o 




Eh 




a 








a 


< 




Cxj 


Eh 






CJ 


<; 






CD 






EH 






















ro 




< 


CD 






< 


(X) 


< 




Cm* 


> 


Eh 


CD CD 




c^ 


CD 




O 


Q 




CD 


CD 








ffi 




CD 


O 


















ffi 




CD 




o 




CJ 






EH 






EH 


Cm CJ 


r- 


CO 


CJ 




CD 


CD 






Eh 


CJ 






EH 






CD 






CJ 


O Eh 






CJ 






Eh : 




CD 


CD 


< CJ 




M 


Eh 






< 






CD 


Ph CD 






< 












< 


CD 






CJ 






EH 




CD 


CD 






EH 


CJ 




CO 


CD 






a 


CJ 






<c 












CD 


O 






o 








CD 


CD 


pc: cd 






Eh 








CD 


u 






Eh 










EH 


CJ 




Cd 


CD 






CD 


CO 


CJ 






CD 




< 


CJ 




EH 


CD 






CJ 






CD 




CD 


CD 






CJ 






Eh 


W 


< 


& CD 




CD 


CD 




O 


CD 




a 


Eh 






CD 






Eh 




CD 


CJ 






CD 






CJ 




Eh 


co CD 










>H 


< 




Eh 


< 






< 






EH 




CD 


CD 






CD 


o 




EH 




Eh 


S Eh 




> 


EH 


CD 




< 




CJ 


< 






CD 


ffi 




EH 








vH 














CD 




c< 




O 








o 


< 




Q 


CO 


CJ 




> 


EH 




a 


CD 


O 




Eh 






CD 








HJ 














CD 


Eh 






CJ 






O 


> 


EH 






Q 


< 




a 




CD 


Eh 






CD 






CD 




CD 


CJ 


O 




< 






CD 


CJ 




CO CD 


CD 


< 


CJ 




Eh 


O 




CD 


< 


ffi 


CD 






< 



rH Q 



E-< 



CO 
CD 
CD 
CD 
CD 
Co 
CD 
CD 
CD 
CD 
CO 



CD 
Eh 

< 

CD 
< 
CJ 

O 
Eh 

< 

CD 

^ 4 
CJ 

CD 
CJ 
Eh 

CD 

P 
CD 

CJ 
CD 
CD 

Eh 
CD 
CD 

CJ 
CD 
CD 

C_> 
CD 

*q 

CJ 
CD 
CD 

Eh 
CD 
CD 

*C 
CD 
CD 

CJ 

CD 
CD 

s 



M 
■ — [ 

rd 
co 



EH 

< 

^ EH 

Eh 

EH 

a 

CO CD 
O CJ 

ro co CD 

Eh 
H Eh 
< 

a 

CO CD 
rf! 

CD 
O < 
CJ 

Eh 
CO CD 

f< a 

CD 

a 

c< CD 
o 

a 
a cd 

EH 

fH CJ 
CJ 

O CJ 
CM EH CJ 
< 

U 
> Eh 
CD 



O 
CD CD 
CD 

Eh 
CO CD 
< 



C^ 



O 
Eh 
Eh 



rH 
PS 
Q 
U 



a 

Cm 1 CD 

o < 
CD CO O 

H^l EH 
< 

Cm CJ 
CJ 

CJ 
> Eh 
CD 

CD 
CD a 
CD 

Eh 
CO CD 
< 

o 

CD 
(-3 Eh 
EH 



CO 



CD 



CJ 
co O 



< a 

CD 



o 

H- 1 



Eh 
CJ 
C.D 



CM 
Cd 
Q 

a 

hH 1 



CJ 



co 



co 



CJ 

< 

EH 

CD 
JSS. 



o CD 

cd o <: 

i—l CJ 



o 



oo 
Cd 
Q 
CJ 



EH 
>h < 
Eh 



CJ CD 
Eh 

CJ 

>H < 
EH 

o 

>H < 
Eh 

Eh 

<; o 

CD 

Eh 
£m Eh 
Eh 

Eh 
Q < 
CD 

CD 

o eh 

oo Cm CJ 
CJ 

a 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



2/17 



FIG. 2 



o 
o 



§5 



O =: 

-C CO 
>< CO 



r 



lac RBS 
promoter 











amber 













leader 



linker peptide 
tag 



plT1/plT2 



-(colE1 orj) 



amp 



gm 



{M13 ori) 



RBS 

CAGGAAACAGCTATGACCATGATTACGCCAAGCTTGCATGCAAATTCTATTTCAAGGAGACAGTCATA ATG AAA TAC CTA 

— > * M K Y L 

LMB3 



Sf il 



Ncol 



TTG CCT ACG GCA GCC GCT GGA TTG TTA TTA CTC GC G GCC CAG CCG GCC ATG GCC GAG GTG TTT 
LPTAAAGLLLLAAQPAMAEVF 

Xhol linker 
GAC TAC TGG GGC CAG GGA ACC CTG GTC ACC GT C TCG AG C GGT GGA GGC GGT TCA GGC GGA GGT 
DYWGQGTLVTVS SGGGGSGGG 



Sail NotI 
GGC AGC GGC GGT GGC GG G TCG AC G GAC ATC CAG ATG ACC CAG GCG GCC GCA GAA CAA AAA CTC 

GSGGGGSTDIQHTQAAAEQKL 
< 

link seq new 

HIS-tag 

CAT CAT CAT CAC CAT CAC GGG GCC GCA 
HHHHHHGAA 
(insertion in pIT2 only) 

my c- tag Gene III 

ATC TCA GAA GAG GAT CTG AAT GGG GCC GCA TAG ACT GTT GAA AGT TGT TTA GCA AAA CCT CAT 
ISEEDLNGAA* TVESCLAKPH 

< ■ 

pHEN seq 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



3/17 



en 

I 



oo 

ES 



CM 



CO 

CD 

Li. 



CNJ 



I 



es 



oo 

CNJ 

1—1 

CO 
CP) 
CO 

r— 

lO 

oo 

CNJ 



cn 



CO 

r— 
lo 

LO 



oo 

CNI 



en 

CO 

lo 

LO 

m 
u 

rd 

CM 
i — I 
CO 
CTi 
CO 

r—- 

wo 

CO 

CNl 



lo 



LO 



CO 



CNJ 



co 

cn 
CO 

lo 



LO 

CO 
CNJ 



cr> 

CO 

lo 

LO 

co 
cd 

CNI 



cn 

CO 

r- 
lo 

LO 

co 

CNJ 
1—1 

CO 

en 

CO 

t — 

LO 



LO 

ro 

CNl 



CO 
CTv 
CO 

t — 

LO 
LO 

co 

CNJ 

4' I 

co 
cn 
co 
t — 

LO 
LO 

oo 

CNl 



cn 
oo 
[ — 

LO 

LO 

ro 

CNJ 



co 
co 

r> 

E-h 
£> 
h-q 
E-t 

CD 
CX 
CD 



r-3 
CD 



o* 

i-h" 

X 
r-— I 

s; 

co 

Q 
PS 
CO 

I — I 

OS 



CD 

t> 

CO 
Q 

£ 

>H 

E-h 
CO 
CD 
CD 
CO 
CD 
CO 



CO 
CO 

g 

CD 
D-i 

OS 



CO 



>H 

CO 



CO 

& 

CD 
CO 

o 

CO 

r-q 

OS 

r-q 

CO 
CD 
CD 
CM 

> 

CD 
CD 
CD 
CO 



I 

r 
i 

Eh 
r-H* 



t 
! 

I 

D-i 
CD 



I 
» 
l 

CD 

CD 
cu 



t 

> 

h-H* 



DS 


CD 


CD 


DS 


OS 


CX 


OS 


CO 


OS 


53 












> 


f=C 


CO 


*C 


CO 


He 




>H 


CO 


HC 


co 




>H 


E-* 


D-. 


co 


E-h 




53 


E-h 




Oh 




CD 




OS 


.< 


J? 




CO 


CO 


CD 


53 


On 




CO 


O-j 


S 


CO 


1 


t 


J 


l 


i 


1 


I 


i 


I 




Q 


PS 


co 




Cx3 


E-h 




E*H 



i 


t 


i 


1 


) 


i 


i 


1 


i 


i 


i 


1 




o» 


:*s 


CO 




r-3 






KV 




fA-L 


53 



CO 



CO 



CO 

ES 



CNJ 

OS 

s 



CNJ 

ES 



QS 

8 



ES 



oo 
r— 

LO 
LO 

ro 

CNl 



CTi 
CO 



LO 



LO 



ro 



CNl 



OO 

0- 

LO 
LO 

ro 

CNl 

1" "I 

CO 
CTv 
OO 

r— 

LO 
LO 

co 

CNJ 



OO 

r— 

LO 
LO 

ro 

CNl 
1 — I 
CO 

en 

CO 



LO 
LO 

oo 

CNJ 



CTv 
CO 
( — 
LO 
LO 

co 

CNI 
r-H 

CO 

cn 
oo 

C- 
LO 
LO 



oo 

CNl 
r-H 
CO 
CTi 
OO 

r— 

LO 
LO 



co 

CNJ 
r-H 
CO 

cr> 
oo 
r— 

LO 
LO 

CO 
CNl 
r-H 
CO 

cn 
oo 
[ — 

LO 
LO 

ro 

CNJ 



os 
i— i 

CO 

5 

E-h 

CD 

o» 



CD 
>h 
>h 
E-h 

o 

CO 
D-i 

o» 

r— -} 
CO 
CO 
r-H 
E-H 
-H* 

g 

E-h 
CD 
CO 
CD 
co 

CD 
co 

Cu 

OS 

CO 

p-4 
^> 

CD 



CO 

co 

CO 



>H 
I— I 

v-O. 

O-i 



CD 
CM 

t*s 

>H 



>H 

CO 

co 

r— f 

CO 
CN 
CO 



o 

E-h 
l — f 
E-h 

:> 
ds 

Q 
CD 
£> 
co 

f=C 
co 

co| 

CO 

o-« 

CO 

s 



DS 



r- 


E-h 


1 


1 


i 


LO 


52: 


CD 


CLi 


r-H* 


LO 


CM 


1 


1 


1 




E-h 


CO 


l-H* 


Q 


ro 


CO 


DS 






CNJ 


>-> 








r— I 


CO 


D-I 






CO 










cn 











DS h-q 

r— J H< 



CO 

ra 

o 
> 



oo 

> X 



CNl 

> 



> 



CD 

15 



CO 













-r-J 


to 


-a 


-a 




CO 


ra 




CO 


CO 


co 


1 | 






f-H 


LO 


LO 


1 1 






g 




CD 


CJ 












PC 










> 


> 


> 



=3 

CO 

^ CO LO CO 
J> IkS PJ CD 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



4/17 



Q 

o 



1.6 
1.4 
1.2 
1 

0.8 
0.6 
0.4 
0.2 
0 



■:■£;:■:•: 



FIG. 4 

Phage ELISA of a dual specific ScFv antibody K8 



[Mliil 



3 4 5 

Antigens 



1- HSA 

2- APS 

3- b-gal 

4- Peanut 

5- BSA 

6- iysosyme 

7- cytochrome c 



FIG. 5 

Soluble ELISA of the Dual Specific ScFv Antibody K8 
2i 




600 



K8 ScFv concentration (nmol) 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



5/17 



o 

O 



1.6 
1.4- 

1.2- 
1 - 

0.8 

0.6 

0.4- 

0.2- 
0 



FIG. 6 

Soluble ScFv ELISA of K8V /dummy V H clone 





1 



2 3 
Antigens 



1- BSA 

2- b-gal 

3- APS 

4- Protein A 



FIG. 7 



RBS 

CAGGAAACAGCTATGACCATGATTACGCCAAGCTTGCATGCAAATTCTATTTCAAGGAGACAGTCATA ATG AAA TAC CTA 
- „> M K Y L 

LMB3 

Sfil Ncol 



TTG CCT ACG GCA GCC GCT GGA TTG TTA TTA CTC GC G GCC CAG CCG GCC ATG GCC GAG GTG TTT 
L P T AAAGIiL L LAA Q P ~ A M A E V F 



> 

Xhol linker 
GAC TAC TGG GGC CAG GGA ACC CTG GTC ACC GT C TCG AG C GGT GGA GGC GGT TCA GGC GGA GGT 
DYWGQGTLVTVS SGGGGSGGG 



Sail NotI 

GGC AGC GGC GGT GGC GG G TCG AC G GAC ATC CAG ATG ACC CAG GCG GCC GC A GAA CAA AAA CTC " 

GSGGGGSTT) IQMTQA ~ A " A £7 Q K L 
< 

link seg new 

HIS -tag 

CAT CAT CAT CAC CAT CAC GGG GCC GCA 
HHHHHHGAA 
(insertion in V domain vector 2 only 



myc-tag Gene III 

ATC TCA GAA GAG GAT CTG AAT GGG GCC GCA TAG ACT GTT GAA AGT TGT TTA GCA AAA CCT CAT 
ISEEDLNGAA*TVESCLAKPH 

< 

pHEN seq 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



6/17 



lac RBS 
promoter 



-^p15 or\y 



FIG. 8 



o 
o 




Is 



leader linker 



CH vector 



o 

CO 



{M13ori> 



^ — cc 




CH gene myc 2x 

tag ochre 



Cm 



FIG. 9 



o 



CO CO 



— II — MB 



lac 
promoter 



RBS leader 



CK vector 



-(colEI orj)- 



amp 



• * • ■ » • » 



CK gene 



CD 
O 

CO 



o 



flag 
tag 




S3 



2x 

ochre 



<M13ori> 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



7/17 



^ 160 



o 
o 



LL. 



FIG. 10 

TNF receptor assay 




0.1 1 10 100 1000 

dAb/dimer concentration nM 



PEP1-5d4 
PEP1-5-19d4 
PEP1-519 monomer 



FIG. 11 



o 
o 



CO 



LL 

z: 



TNF receptor assay 




1 10 100 

dimer concentration nM 



1000 



-x- 



PEP1-5d1 
PEP1-5d2 
PEP1-5d3 
PEP1-5d4 
PEP1-5d5 
PEP1-5d6 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



8/17 



PCT/GB2003/002804 



FIG. 12 

TNF Receptor assay 



110 




-10-J 1 

0.1 1 10 100 1000 



dAb/dimer concentration nM 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



9/17 



PCT/GB2003/002804 



FIG. 13 



Dummy V H sequence for library 1 



1 


E 
GAG 
CTC 


V 
GTG 
CAC 


Q 
CAG 
GTC 


L 
CTG 
GAC 


L 
TTG 
AAC 


E 
GAG 
CTC 


S 
TCT 
AGA 


G 
GGG 
CCC 


G 
GGA 
CCT 


G 
GGC 
CCG 


L 
TTG 
AAC 


V 
GTA 
CAT 


Q 
CAG 
GTC 


P 
CCT 
GGA 


G 
GGG 
CCC 


G 
GGG 
CCC 


49 


S 
TCC 
AGG 


L 
CTG 
GAC 


R 
CGT 
GCA 


T i 
CTC 
GAG 


S 
TCC 
AGG 


C 
TGT 
ACA 


A 
GCA 
CGT 


A 
GCC 
CGG 


S 
TCC 
AGG 


G 
GGA 
CCT 


F 
TTC 
AAG 


T 
ACC 
TGG 


F 
TTT 
AAA 


S 
AGC 
TCG 


S 
AGC 
TCG 


Y 
TAT 
ATA 


97 


A 
GCC 
CGG 


M 
ATG 
TAC 


S 
AGC 
TCG 


W 
TGG 
ACC 


V 
GTC 
CAG 


R 
CGC 
GCG 


Q 

CAG 
GTC 


A 
GCT 
CGA 


P 
CCA 
GGT 


G 
GGG 
CCC 


K 
AAG 
TTC 


G 
GGT 
CCA 


L 
CTA 
GAT 


E 
GAG 
CTC 


W 
TGG 
ACC 


V 
GTC 
CAG 




S 


A 


I 


S 


G 


S 


G 


G 


S 


T 


Y 


Y 


A 


D 


S 


V 


145 


TCA 


GCT 


ATT 


AGT 


GGT 


AGT 


GGT 


GGT 


AGC 


ACA 


TAC 


TAC 


GCA 


GAC 


TCC 


GTG 




AGT 


CGA 


TAA 


TCA 


CCA 


TCA 


CCA 


CCA 


TCG 


TGT 


ATG 


ATG 


CGT 


CTG 


AGG 


CAC 


193 


K 
AAG 
TTC 


G 
GGC 
CCG 


R 
CGG 
GCC 


F 
TTC 
AAG 


T 
ACC 
TGG 


I 

ATC 
TAG 


S 
TCC 
AGG 


R 
CGT 
GCA 


D 
GAC 
CTG 


N 
AAT 
TTA 


S 
TCC 
AGG 


K 
AAG 
TTC 


N 
AAC 
TTG 


T 
ACG 
TGC 


L 
CTG 
GAC 


Y 
TAT 
ATA 


241 


L 
CTG 
GAC 


Q 
CAA 

GTT 


M 
ATG 
TAC 


N 
AAC 
TTG 


S 
AGC 
TCG 


L 
CTG 
GAC 


R 
CGT 
GCA 


A 
GCC 
CGG 


E 
GAG 
CTC 


D 
GAC 
CTG 


T 

ACC 
TGG 


A 
GCG 
CGC 


V 
GTA 
CAT 


Y 
TAT 
ATA 


Y 
TAC 
ATG 


C 
TGT 
ACA 




A 


K 


S 


Y 


G 


A 


F 


D 


Y 


W 


G 


Q 


G 


T 


L 


V 


289 


GCG 


AAA 


AGT 


TAT 


GGT 


GCT 


TTT 


GAC 


TAC 


TGG 


GGC 


CAG 


GGA 


ACC 


CTG 


GTC 




CGC 


TTT 


TCA 


ATA 


CCA 


CGA 


AAA 


CTG 


ATG 


ACC 


CCG 


GTC 


CCT 


TGG 


GAC 


CAG 


337 


T 
ACC 
TGG 


V 
GTC 
CAG 


S 
TCG 
AGC 


S 
AGC 
TCG 



























SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



10/17 



PCT/GB2003/002804 



FIG. 14 



Dummy V H sequence for library 2 



1 

1 


E 
GAG 
CTC 


V 
GTG 
CAC 


Q 
CAG 
GTC 


L 
CTG 
GAC 


L 
TTG 
AAC 


E 
GAG 
CTC 


S 
TCT 
AGA 


G 
GGG 
CCC 


G 
GGA 
CCT 


G 
GGC 
CCG 


L 
TTG 
AAC 


V 
GTA 
CAT 


Q 
CAG 

GTC 


P 
CCT 
GGA 


G 
GGG 
CCC 


G 
GGG 
CCC 


49 


S 
TCC 
AGG 


L 
CTG 
GAC 


R 
CGT 
GCA 


L 
CTC 
GAG 


S 
TCC 
AGG 


C 
TGT 
ACA 


A 
GCA 
CGT 


A 
GCC 
CGG 


S 
TCC 
AGG 


G 
GGA 
CCT 


F 

TTC 
AAG 


T 
ACC 
TGG 


F 
TTT 
AAA 


S 
AGC 
TCG 


S 
AGC 
TCG 


Y 
TAT 
ATA 


97 


A 
GCC 
CGG 


M 
ATG 
TAC 


S 
AGC 
TCG 


W 
TGG 
ACC 


V 
GTC 
CAG 


R 
CGC 
GCG 


Q 
CAG 
GTC 


A 
GCT 
CGA 


P . 
CCA 
GGT 


G 
GGG 
CCC 


K 
AAG 
TTC 


G 
GGT 
CCA 


T i 
CTA 
GAT 


E 
GAG 
CTC 


W 
TGG 
ACC 


V 
GTC 
CAG 




S 


A 


I 


S 


G 


S 


G 


G 


S 


T 


Y 


Y 


A 


D 


S 


V 


145 


TCA 


GCT 


ATT 


AGT 


GGT 


AGT 


GGT 


GGT 


AGC 


ACA 


TAC 


TAC 


GCA 


GAC 


TCC 


GTG 




AGT 


CGA 


TAA 


TCA 


CCA 


TCA 


CCA 


CCA 


TCG 


TGT 


ATG 


ATG 


CGT 


CTG 


AGG 


CAC 


193 


K 
AAG 
TTC 


G 
GGC 
CCG 


R 
CGG 
GCC 


F 
TTC 
AAG 


T 
ACC 
TGG 


I 
ATC 
TAG 


S 
TCC 
AGG 


R 
CGT 
GCA 


D 
GAC 
CTG 


N 
AAT 
TTA 


S 
TCC 
AGG 


K 
AAG 
TTC 


N 
AAC 
TTG 


T 
ACG 
TGC 


L 
CTG 
GAC 


Y 
TAT 
ATA 


241 


L 
CTG 
GAC 


Q 
CAA 
GTT 


M 
ATG 
TAC 


N 
AAC 
TTG 


S 
AGC 
TCG 


L 
CTG 
GAC 


R 
CGT 
GCA 


A 
GCC 
CGG 


E 
GAG 
CTC 


D 

GAC 
CTG 


T 
ACC 
TGG 


A 
GCG 
CGC 


V 
GTA 
CAT 


Y 
TAT 
ATA 


Y 
TAC 
ATG 


C 
TGT 
ACA 




A 


K 


S 


Y 


G 


A 


X 


X 


X 


X 


F 


D 


Y 


W 


G 


Q 


289 


GCG 


AAA 


AGT 


TAT 


GGT 


GCT 


NNK 


NNK 


NNK 


NNK 


TTT 


GAC 


TAC 


TGG 


GGC 


CAG 




CGC 


TTT 


TCA 


ATA 


CCA 


CGA 


NNK 


NNK 


NNK 


NNK 


AAA 


CTG 


ATG 


ACC 


CCG 


GTC 


337 


G 
GGA 
CCT 


T 
ACC 
TGG 


L 

CTG 
GAC 


V 
GTC 
CAG 


T 
ACC 
TGG 


V 
GTC 
CAG 


S 
TCG 
AGC 


S 

AGC 
TCG 



















SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



11 I M 



PCT/GB2003/002804 



FIG. 15 



Dummy V K sequence for library 3 

DIQMTQSPSSLSASVG 
1 GAC ATC CAG ATG ACC CAG TCT CCA TCC TCC CTG TCT GCA TCT GTA GGA 
CTG TAG GTC TAC TGG GTC AGA GGT AGG AGG GAC AGA CGT AGA CAT CCT 

DRVTITCRASQS I S S Y 
4 9 GAC CGT GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC ATT AGC AGC TAT 
CTG GCA CAG TGG TAG TGA ACG GCC CGT TCA GTC TCG TAA TCG TCG ATA 

LNWYQQKPGKAPKLL I 
97 TT A AAT TGG TAC CAG CAG AAA CCA GGG AAA GCC CCT AAG CTC CTG ATC 
AAT TTA ACC ATG GTC GTC TTT GGT CCC TTT CGG GGA TTC GAG GAC TAG 

Y_AASSLQSGVPSRFSG 
14 5 TA T GCT GCA TCC AGT TTG CAA AGT GGG GTC CCA TCA CGT TTC AGT GGC 
ATA CGA CGT AGG TCA AAC GTT TCA CCC CAG GGT AGT GCA AAG TCA CCG 

SGSGTDFTLTISSLQP 
193 AGT GGA TCT GGG ACA GAT TTC ACT CTC ACC ATC AGC AGT CTG CAA CCT 
TCA CCT AGA CCC TGT CTA AAG TGA GAG TGG TAG TCG TCA GAC GTT GGA 

EDFATYYCQQ S Y S T P N 
2 41 GAA GAT TTT GCT ACG TAC TAC TGT CAA CAG AGT TAC AGT ACC CCT AAT 
CTT CTA AAA CGA TGC ATG ATG ACA GTT GTC TCA ATG TCA TGG GGA TTA 



TFGQGTKVEIKR 
2 89 ACG TTC GGC CAA GGG ACC AAG GTG GAA ATC AAA CGG 
TGC AAG CCG GTT CCC TGG TTC CAC CTT TAG TTT GCC 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



12/17 



FIG. 16 

Nucleotide and amino acid sequence of anti MSA dAbs MSA 16 and MSA 26 

A: MSA 16 

GAC ATC CAG ATG ACC CAG TCT CCA TCC TCC CTG TCT GCA TCT 
DIQMTQSPSSLSAS 

GTA GGA GAC CGT GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC 
VGDRVTITCRASQS 

ATT ATT AAG CAT TTA AAG TGG TAC CAG CAG AAA CCA GGG AAA 
1 I K H L K W ... YQQKPGK 

GCC CCT AAG CTC CTG ATC TAT GGT GCA TCC CGG TTG CAA AGT 
APKLLIYGASRLQS 

GGG GTC CCA TCA CGT TTC AGT GGC AGT GGA TCT GGG ACA GAT 
GVPSRFSGSGSGTD 

TTC ACT CTC ACC ATC AGC AGT CTG CAA CCT GAA GAT TTT GCT 
FTLTISSLQPEDFA 

ACG TAC TAC TGT CAA CAG GGG GCT CGG TGG CCT CAG ACG TTC 
TYYCQQGARWPQ TF 

GGC CAA GGG ACC AAG GTG GAA ATC AAA CGG 
GQGTKVEIKR 



B: MSA 26 

GAC ATC CAG ATG ACC CAG TCT 

D I Q M T Q S 

GTA GGA GAC CGT GTC ACC ATC 

V G D R V T I 

ATT TAT TAT CAT TTA AAG TGG 

I Y Y H L K W 

GCC CCT AAG CTC CTG ATC TAT 

A P K L L I Y 

GGG GTC CCA TCA CGT TTC AGT 

G V P S R F S 

TTC ACT CTC ACC ATC AGC AGT 

F T L T I S S 

ACG TAC TAC TGT CAA CAG GTT 

T Y Y C Q Q V 

GGC CAA GGG ACC AAG GTG GAA 

G Q G T K V E 



CCA TCC TCC CTG TCT GCA TCT 
P S S L S A S 

ACT TGC CGG GCA AGT CAG AGC 
T C R A S Q S 

TAC CAG CAG AAA CCA GGG AAA 
YQQKPGK 

AAG GCA TCC ACG TTG CAA AGT 
K A S T L Q S 

GGC AGT GGA TCT GGG ACA GAT 
G S G S G T D 

CTG CAA CCT GAA GAT TTT GCT 
L Q P E D F A 

CGG AAG GTG CCT CGG ACG TTC 
R K V P R T F 

ATC AAA CGG 
I K R 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



14/17 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

15/17 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



16/17 



(a) 
0.35 

0.3 
0.25 

0.2 
0.15 

0.1 
0.05 
0 



FIG. 19 



m Series 1 



TAR 1-5- TAR 1-5- 
19dAb 19dAb 
+ MSA 



Dual Dual 
specific specific 

+ MSA 



(b) 



biotinylated anti-TNF 



TNF 





chromogenic 
substrate 



Streptavidin-HRP 



TNFRI/Fc chimera 



Anti-Fc capturing antibody 



(c) 



100 



~ 80 



o 
o 



CO 



LL 



60 



40 



5 20 



0 



-20 



't*t*t i * *lal 



LH 



TNF Receptor assay 



LH + 
MSA 



1 nnnnnnnno" 



TNF + 
MSA 



MSA 
only 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



17/17 



PCT/GB2003/002804 



FIG. 20 

TNF Receptor assay 
120 n 



100 



o 
o 



CD 

• HAS 

_Q 
Ll_ 




80 



60 




18nM LH + MSA 
MSA 

18nM LH + HSA 
HSA 



Serum Albumin (mg/ml) 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



1 



PCT/GB2003/002804 



CD 



CO 



LU 

U 

LU 

C? 

LU 

CO 



Eh 

n < 

CD 

eg 

CD 
E-J 

g 

CJ 
CO CD 



Eh 

CD 
CD 

IS 

fa Eh 
Eh 
f=C 

CD CD 
CD 
a 

CO O 

Eh 

u 
a 

CD 

j< CJ 
CD 
Eh 

O CD 
Eh 
U 

CO u 
Eh 

O 

^B 

Eh 
OS CD 
O 
CD 

B 

CJ 

CO CJ 
Eh 
CD 
CD CD 
CD 
CD 
CD CD 
CD 
Eh 
CJ 



ft 



o 

CD 

its 

CD 
CD 
Eh 
Eh 



CJ 

u g 

CD 
CD CD 
CD 
Eh 

CO CJ 

s 

CD 
h3 Eh 
Eh 
CD 
Eh 



Of 



CJ 

8 



CD 
> Eh 
CD 
CD 



w w < 



Pi CD 
CJ 
CJ 

CD CD 
CD 



CD 



CD 
Eh 



CD 
O 
CO CJ 
Eh 

a 

«S 
CD 

CJ 

>* & 
Eh 
CJ 

Eh 

EH U 

0 

CO CD 
CD CD 

8 

CO CD 
CJ 

ft 

u 
u 

CD CD 
CD 
Eh 

>* < 
Eh 
Eh 

H Eh 



a 
to CD 

CD 

co a 

Eh 

a 

> EH 

8 

EH CJ 

> EH 

CD 
CD 
i4 Eh 
CJ 
CJ 
U 

Eh 



CD CD 
CD 
CD 

a < 

CJ 
Eh 
CD CD 
CD 

CD 
CD 
Eh 
Eh 
Eh 
Eh 
CJ 
CD CD 
CD 
CD 



fa 



fa 



CD 



.J Eh 
CJ 
U 

ft CJ 

a 
q 

fa 

CD 
CD 

u 

CO 

EH 

1-3 Eh 
CJ 
CD 

ft CJ 

a 

CD 

fa 

CD 



2 g 
■a 

u 

Eh 



Eh 

cj 

Eh CJ 

6 

fa E-H 
Eh 

CD CD 
CD 
CJ 

co a 

Eh 

CJ 
U 



in 

CM 

s 

Eh 



u 

s 

CJ CD 
Eh 
U 

CO U 
Eh 

CJ 

hH* EH 

CJ 
Eh 

p$ cd 

CJ 
CD 
Eh 
CJ 
CJ 

CO CJ 

g 

CD CD 

8 

CD CD 

a 

ft 

cj 

CD 

eg 

> § 

CD 
CD 

Eh 

CJ 
CD CD 
O 
f< 

CD CD 
CD 
CD 

CD CD 
CD 
Eh 

CO CJ 
Eh 
CD 

CD 

a eh 

EH 

CD 

CJ 
CD 
a < 

CJ 
CD 

> Eh 
CD 
CD 

w 

CD 



CtJ CD 
O 
CJ 

CD CD 





EH 


> 






CD 




CJ 


CO 


O 




EH 






Q 






9 






< 


O 


CD 






>* 










I 


EH 










EH 




B 


fa 


CD 










Eh 


CD 


CD 




CD 




El 




CJ 


EH 










Of 


6 






CO 


CD 










H 






s 



fa 



fa 



O 
CO CD 



CO o 

B 

CD 
U 
H CJ 

> 3 

hH" EH 

O 

o 

E-> CJ 



a 



8 
6 

CJ 



O CD 
O 
O 

& O 
Eh 
O 

Eh 

2 




fa 



fa 



EJ 

| 
Eh 
Eh 
Eh 
Eh 

CD 
CD 



Eh 

> ^ 
CD 

CJ 

CO CD 

CD 

a < 
o 

Eh 
CO CD 

3 

rfj CJ 



fa 



CD 



CJ 
U 
CJ CD 
Eh 
Eh 
Eh CJ 



H Eh 
<! 

O 
Eh CJ 
< 

a 

> Eh 
CD 
Eh 

* 8 



R 



fa 



a 



H 



Of 



a 



o 

CJ 
CD CD 
CD 



fa 



O 
Eh 
Eh 
CD 



Eh U 

Eh 
fa CD 
U 
Eh 
fa U 
O 
CD 



O 
CD 



Eh 

fa Eh 

Eh 

CD 



CD 

a 



o 
cj 

EH 

U CD 
Eh 
O 

Eh 

CJ 

Eh 
CD 
Eh CJ 

IS 

U 

CD 
Eh 
fa Eh 
Eh 
E-J 

O 



fa 



CD 



Eh 

ft CJ 
CJ 

O . 

o 
o 

* CJ 
Eh 

CO O 



CO CD 

a 
s 

EH 

CJ 

EH 

Eh 
Eh 



fa 



Eh 

CD 
CD CD 

CD 
• Eh 



in 



o 



CN 



in 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



10 



to 



cd 



CO 



cn 



en 



a a 

CD 



CO 


u 








Eh 


Pn 


CD 






EH 




CD 


a 




a 






a 




CD 


53 




CJ 




CD 


CD 










CD 


CD 






CD 






co 


CJ 




CD 






CD 


>H 


g 




EH 




3 


<y 


CD 


CD 




Eh 




CJ 






CD 




CD 


> 


EH 










EH 


IS 


CD 




CD 




CD 






CJ 




EH 




CD 




H 




ft 












> 


CD 






U 




S3 




CD 










w 


< 




Eh 




a 


>H 


a 






CD 




52 


CO 


CJ 








Eh 


w 






Eh 








Eh 




52 




a 




> 


Eh 




Eh 






Q 








CD 




O 








53 






a 


Eh 


% 




CJ 


< 




Eh 




EH 








Eh 


a 






EH 




a 




CD 


CD 












CD 


CD 










a 


Pn 


Eh 






CJ 




CD 


CD 






« 


52 


>H 


n 




53 




< 








W 






CD 


CD 










CD 


a 


CD 




CD 








CD 




O 


CD 


CD 










CD 


CO 


a 




52 






CO 


CD 


CD 




EH 






EH 






a 




a 


ft 












Eh 


< 


o 




CJ 












52 








S3 




CO 


CJ 




Eh 


CO 




Eh 


< 




CJ 










S3 


CJ 


a 




Eh 




W 




CD 




CD 


CD 


CD 


a 






ex < 




CD 






CD 


a 


CD 




CJ 




EH 




k3 






Eh 




CJ 


CD 


CD 




PS 




CJ 


CD 




CD 






CD 


CO 


CJ 




CJ 




Eh 


CO 




Eh 




Eh 




CJ 




CD 
















CO 








CJ 




U 


> 


EH 








CD 




Eh 




CD 






H 






CJ 




CD 


CD 


CD 






a 




H 




CD 




CD 






a 




CD 




Eh 




Eh 


Eh 


> 


Eh 


CJ 




Eh 


CO 


52 






CD 




CD 


CD 


CD 










S3 




Eh 




CD 




Eh 






<< 




a 




CD 


H 






a 


♦ 


CJ 


• 


EH 




< 


• 




rH 




rH 


• 


rH 




H 










IT) 




O 


















rH 




rH 





CD > 



H 
o 

CN 



CJ O < 



H CD CD 



a q rfj 



CJ Ph CD 



H 

in 

CM 



H 
o 

CO 



CJ 
CO CD 

< 

CD 
CO CJ 

Eh 
. CJ 
> H 

CD 
• CJ 



i— I 
in 
m 



U 4-1 



to 

n 



m 



< En 

q O u cn 

< E-> 
cd a 

O U U >h 

rtj Eh 

Eh < 

> CD U H 

Eh i< 

a cd 



CO Eh 

s 

Eh 
U 

CO Eh 
CD 
Eh 

^ U 



o 

u i-h" 

pal 

o 



3 



Eh 
O 
Eh 
CJ 
CJ 

Eh 



CO 



CO 



Pit 



CD 

o 

CD 

E-t 

CD 

CD CD 



U CD 

cn Eh <J 

CD CJ 

< EH vJ 

Of O CD W 

O CD 

U CD 

EH < EH 



H 
l 

m 
i 

H 



Eh Q 



S < H 
CD U 
Eh 

O O CD 
O CD 

h rf h 

U CD 

CD U 



*0 



o 

C4 



IT5 



SUBSTITUTE SHEET (RULE 26) 



o 

CO 



o 



WO 2004/003019 



3 



PCT/GB2003/002804 



u 


CD 








Eh 


< 


o 




Eh 


< 


EH 






Eh 


CD 


U 






CD 


CD 




cd 




CD 


H 








a 


CD 






§ 


u 


s 




fa 








cd 




EH 






cj 




EH 


t 






CD 








Eh 




8 










CD 


u 








a 




fa 


Eh 


i 








Eh 




EH 




H 




Eh 






H 




Eh 






CJ 


o 


ID 


s 




CJ 


CD 


U 


CD 




Eh 






ps 


CD 


u 


6 


EH 












a 


CD 


Eh 


< 




CD 


U 


< 


H 




CD 


CJ 


CJ 


CD 




Eh 




cj 


CD 


> 


CD 




u 






Eh 


6 


Eh 






cd 




CD 


u 


> 


Eh 




CD 


o 




GT 




CD 


u 








CD 




Of 




CJ 


Eh 






< 




o 


u 




CJ 


CD 



Of rf Eh 

rf Eh 

CJ CD 

CJ Eh < 

CD U 

Eh < 

CJ CD 

>h 

ril Eh 

>h U CD 
Ei 



< U CD 



CD U 



fa 



U CD 

CD CJ 

CJ CD 

< Eh 

Eh 
CJ CD 



CD CJ 
Eh *C 



*3 




O f< Eh 

O CD 

Eh < 

co cd o 

< Eh 

r< O CD 

CD CJ 

CD CJ 

CD CJ 




f$ Eh 

< O CD 

CD CJ 

Eh < 

CO CJ CD 

Eh & 

CD CJ 

Eh < 

CJ CD 

CJ CD 

CQ CJ CD 

Eh < 

CJ CD 

CO CJ CD 

Eh < 

*C Eh 

CJ CD 

CJ CD 

Eh f$ 



CD ft CJ CD 
CJ CD 
Eh 

CJ CD 



CD CJ 

PS CD CJ 

CJ CD 

< EH 




H 
P 



U3 



(l) 

-H 
H 

P 
in 



LO 



u 



•H 
H 
P 

m 



i 

o 



CD CJ 

t$ Eh 

h3 Eh rf 

Eh < 

CD .CJ 

< CJ CD 

CD U 

Eh rf 

Q < E-J 

CD U 



CO CJ CD 

eh <: 

< Eh 

CD CD CJ 

CD CJ 

CO CD CJ 

CD CJ 



rf Eh 

a< eh 

U CD 

CJ CD 

CD CD CJ 

CD CJ 

CJ CD 
Eh 

Eh ^ 

CD CJ 



CJ CD 
< E-> 



fa 




Of < Eh 

O CD 
£h 

CO CD CJ 
Eh 

j < Eh 

^ U rj 

CD U 

CD CJ 

CD O 

PS 

O CD 

CJ CD 

CJ CD CJ 

Eh e£ 
Eh 

CJ CD 

< Eh 
CJ CD 
Eh 

< Eh 



U CD 

O CJ 

< Eh 

CJ CD 

O CJ 

Eh rf 

EH a 



H 



CJ CD 

Eh CJ CD 
Eh 

CJ CD 

> ^ ^ 
CD CJ 

Eh < 

PS CD O 

O CD 

CJ CD 

Q f< Eh 

CD CJ 

< Eh 

O CD CJ 

CD CJ 

> Eh 




Eh < 

CD CJ 

PS CD CJ 

CJ O 

CJ CD 

CO CJ CD 

§ Eh 

rtj 6 CD 

CD U 



R 

U CD 

J* £JJ EH 

Eh < 

CD U 

Eh CJ CD 

< Eh 

Eh ft 

U O 



CO 



CD CJ 
Eh < 
CJ CD 



Eh 
CD 

CD O 

Eh < 

CO CJ CD 

Eh rsfi 

CD O 

^ Eh < 

O CD 

CJ CD 

CO CJ CD 

Eh < 

O CD 

CO CJ CD 

Eh rtj 

< Eh 

Pi U CD 

CJ CD 

EH rtj 

CO CJ CD 
Eh 

CD U 

Of < Eh 

O CD 

CJ CD 

E-r CJ CD 

fiC Eh 

CD CJ 

EH f< 



< Eh 
CD O 

Eh 
O CD 
CJ CD 
Eh < 

Eh 
CJ CD 

< Eh 
CD O 



Of 




CD 



o o 



6 



H 



fa 



PS 



CD 
O CJ 

CD CJ 

Eh < 

O CJ 

< Eh 



CJ CD 



Pj 



H 

Eh 
> 
Pi 



U CD 

CJ CD 

»3 E-< 

O CD 

Eh <: 

CD O 

B < 

O O 

O CD 

CJ CD 



< Eh 
CO U CD 

Eh < 

< Eh 
CJ O 
U CD 
O CD 

> Eh 

CD CJ 

CD O 

O CD CJ 

CD CJ 

Eh f$ 

CO CD O 

< Eh 
*C Eh 

Of Eh 

U CD 

CD CJ 

Eh *C 



PS O O 

CJ CD 

O O 

CO CJ CD 

< Eh 

< O CD 

CD O 



H 



Eh < 
rt^ Eh 
CJ CD 
CD CJ 
Eh < 
U -CD- 
En < 
CD CJ 
Eh rtj 
U CD 



CO 



^ Eh 

O CJ 

< Eh 
O CD 
CJ CD 
Eh < 

< Eh 
CJ CD 

< H 
CD CJ 



I 

U 

i 



in 
cn 



o 

cn 



SUBSTITUTE SHEET (RULE 26) 



o 



WO 2004/003019 



4 



PCT/GB2003/002804 



cd 



to 



CD 



CJ CD 
H ^ O 
paj Eh 
CD CJ 

C5 U C5 CD U 
o CJ 




£h < 

O CJ 
Eh 

CJ CD 

CD CJ 

CD CJ 

Eh <3 



fa 



CD 



fa 



CD 



03 



CD 



to 



H 



CD 



CD 



CD 



03 



CJ CD 



CD CJ 

a ^ Eh 

CJ CD 

Of < Eh 

CJ CD 

H <3 

CJ CD CJ 

Eh < 

CJ CD 



W < Eh 

U CD 

CJ CD 

< % 
S3 

Eh CJ CD 
Eh 



Eh 



CJ CD 




Em 



Eh rf 

CM CJ CD 

CJ U 
Eh 

a 3 eh 

CJ CD 

CD CJ 

a eh 

CJ CD 

Eh rij 

CO CD a 

rij Eh 

CJ CD 

CQ CD CJ 

< Eh 

a cd 

Eh < 

ss 

CJ CD 



H 



f3 Eh 

U CD 

Eh (3 

CJ CD 

Eh < 

CJ CD 

SH 
CD 

Eh 
Eh 



CD CJ 

CD CJ 

CJ CD 

< Eh 



M 



EH ■< 

P < Eh 

CD CJ 

fd| Eh W 




Eh CJ CD 

<1 Eh 

CD a 

CD CD CJ 

CD CJ 

Eh < 



CD CJ 

CD a 
Eh 

< Eh 
CJ CD 

Eh CJ CD 

< Eh 
CD O 

CD CD CJ 

cd a 



CD 



CD CD 
CD 



3 



CQ 



cd a 

Eh < 



CJ CD 

Eh rfj 

*C Eh 

CD U 



fa 



fa 



CU 



cd en cj cd 

CD CJ CD 

rtj Eh 

cd a 2 H 

CJ CJ CD 



CD CQ 

CD 

CD 

CJ CQ 



CD CD 
CD CD CD 
< CD 



H Eh 



Eh rij 

CD CJ 

< Eh 

CJ CD 

CD CJ 

*d Eh 

U CD 

CJ CD 



, CD CJ 

P4 CD CJ 

CJ CD 

< EH 



H 



<J CD CJ 
Eh CX < Eh 



Eh 

Eh rtj 



H 



C9 CJ 

rfj Eh 

Eh CJ O 

< Eh 

CD CJ 

CD CD CJ 

CD CJ 
Eh 



Eh U CD 

*C EH 

CD U 

CD CD O 

CD CJ 



S 



6 



CD CO CJ CD 

< Eh < 

CJ ^ Eh 

EH CM CJ CD 



fa 



3 



Of 



CO 



8 



0 



H 



U CD 
CJ CD 

cj a 

CD CJ 
Eh 



fa 



Eh rfj 

Oi CJ CD 

CJ CD 

sfi Eh 

Of f3 Eh 

CJ CD 

O CJ 

^ Eh ril 



CO CD O 

riJ Eh 

CJ CD 

to CD CJ 

< Eh 

CJ CD 

Eh < 



Of 



O 



l-l 



H 



CD CD CD CJ 

*C CD CJ 

3 

Eh , H 

CD « 2 Eh 

CD < Eh 

CD CD CJ 

Eh Of Eh 

O U CD 

rf{ CD CJ 



rf! Eh 
U CD 



t£ Eh 
CJ CD 
CJ CD 



, O CD 

Ph CD CJ 

U CD 

< Eh 



H 



fa 



fa 



rfl EH 
2! Eh 



Of 



Pi 



to 



to 



Of 



CD 



EH 



U 



fl> Of rij 



u 

i 

3 



1 

H 

9 



U 
(d 

Cm 



to 



O 



to 



O 



to 

CO 



o 



to 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



5 



PCT/GB2003/002804 



u cd 

EH fit <y 

fit En 

cd cj 

CD U CD O CD 

Eh rtj 




CD CJ 

fit Eh 

CJ CD 

CD CJ 




CJ CD 



CD CJ 

CD CJ 

CJ CD 

U 5 

CD CJ 

H CJ CD 

fit Eh 

CD CJ 

Eh fit 

CD CJ 
Eh 

U CD 

CJ CD 



Eh 
H Eh 

fit Eh 
U CD 
Eh < W 
pjj Eh 
CJ CD 

CJ CD Of CD CJ 
fit EH 



a cd 



CJ CD 

Eh fit 

CD CJ H 

CD CJ 



CJ CD 
Eh 



Eh 
fit Eh 



CD CJ 
CD CJ Of CD CJ 
rfj Eh 




Of 



CJ CD 

fit Eh 

< Eh 

O CD 



U CD CJ Eh gC 



CD CJ 
Eh < 



Eh 

CD CJ 

EH 

CJ CD 

CJ CD 
Eh 

riS Eh 

CJ O 

CD CJ 



Eh < 

cj CD 

CD CJ Pn 

Eh rf 

i*C Eh 

Eh fit Q 

O CD 

Eh < 

^ Eh W 

CD CJ 



CD O 

g Eh 

Eh fit 

!h U O 

Eh fit 

EH CD CJ 

CJ CD 

Eh < 

rtj CJ CD 



CD CJ 
Eh 



6 



rij H O Eh 
CJ CD fa O CD 
Eh fit 



CD U 



P4 



fa 




EH 



CD O W 

Eh < Eh 

O O 

fit Eh Eh 

CD CD CJ 

CD CJ 

CD CJ CD 

Eh fit 



3 




Eh 

rtj £H 



O CD CO CJ CD 

£H tfj a 

fit S O CD 

CD CD CJ U CD 

CD CJ CD CD O 



Eh 

CO CD O 

fit Eh 

^ Eh 

rt^ CJ CD 

CD CJ 

CD CJ 

CD CJ 



fit Eh 
CD CO U CD 
fit 



Pi 



Eh & fit O CD 

fit Eh CD CJ 

to O CD Eh < 

cj a cd cj 

O CD fit E-» 

Eh 2 CD U 

"8 a 6 ' 



Eh 
CD 



CJ CD O CD CJ 




O CJ 

CJ Of fit Eh 

U CD 

fit Eh 

Eh Of < Eh 

Eh O CD 

H Eh fit 

CD O O O 

Eh fit 

CJ CD 



EH 



fit 



CJ CD 

^ Eh 

Eh fit 

CD O 

EH fit Eh CJ p 

Eh fit E-J 

U CD Eh ^ 

U CD 



U 

■a 

t-i 



a fit Eh O CJ 

CD CJ Eh CJ CD 

*U Eh 

CD CD O 

CD O Jh 

^ ?J Eh 

> Eh rij 

CD CJ H 

Eh rf 

CJ CD 

CO 

jr-J rf ^ 

CD O ^ 

*1 ^ 

CO CJ CD 

Eh ri^ W 

CD CJ 

EH 

CJ CD 



O CD 
CO CJ CD 



Eh i< rtj U 



S3 „ 



CJ CD 

CO CJ CD 
Eh 

riJ Eh 

U CD 

U CD CD 

Eh fit 



•d co u CD 
o 

CN 

I 

H 




CD O 
CD CJ 
CJ CD 



I 

Eh 

cs 

•rl 



Of Eh 

CJ CD W 

CJ CD 

Eh CJ CD 

fit Eh 

CD U 

. Eh fit 



fit H 
0* CJ o 

o o 




a fit Eh 
CJ CD fa 
CD O 



Eh 
CD U 



O ft Ej 



Q) Ol fit Eh 

I 



CJ CD Q rf) 
U CD 



H 



CJ CD fn rtj Ej 
CJ CD Eh fit. 

eh cd a 



EH 
CJ CD 



CD CJ 
Eh 

b § 

< Eh 




CJ CD CO CJ CD 

< eh H si: a 

Eh fit E-J 

Eh 21 CD CD CJ 




Eh 

a o 



CJ CD 
CD CJ CD CD CJ 



CD CJ 
Of fit Eh 



Eh < 

CO CD O 

fit Eh 

■3 EH 

i< O O 

CD CJ 

CD CJ 

CD CJ 




CJ CD CO CJ CD 



CM CJ CD 

O CD CD 

U CD 

CD O 



U CD 
Eh 



Eh 

U CD 



O O O CD CJ 
U CD 
CJ CD CJ 




cd a Of fit 

Eh < 



a 



CD O 
' Eh 



O CD CJ CD CJ 




Pi CD u 



< Eh 
Eh 

U CD 

< &H 
Eh fit 
O U 
CJ CD 



CJ CD <I CJ CD 
O CD CD CJ 



fit 
Eh 



Q ri! Eh 

CD O 

fit Eh 

CD CD O 

CD O 

fit Eh 

CJ CD 

CJ CD 



CD O 

< Eh 
Eh 

Eh g 

^ Eh 

Eh < 

a cd 

Eh rf 

fit Eh 

CD CJ 



fx] 



CJ CD 




CO 



fit 



fit Eh 
CJ CD 
CD CJ 



^ Eh fit 



EH 



EH < 

CO CJ CD 

Eh rf W 

CD O 



CJ CD Ch CJ CD 

CJ CD CJ CD 

Eh fit < Eh 

O CD Of fit Eh 

CD CJ CJ CD 
Eh 




U 

CD 

•P 
Jh 
ni 

Pi 



CO U CD 

EJ fit 

fit Eh 

CD CD CJ 

CD CJ 

Eh fit 

CO CD CJ 

fit Eh 

CJ CD 

CD CJ 



H 



O O 



Of 



H 



CJ CD 

CO CJ CD 

Eh fit 

fit Eh 

rij CJ O 

CD CD CJ 

H Eh fit 

CJ W < Eh 



CD CJ 



g 



2 



Of 



HI 



CJ 
CJ 

Eh 

ft 
CD 

s 

CJ 
CD 



Eh 0 <J Eh 
U O CD 
fit CD U 



Of 



£h 
O 
Eh 

CD >h 



Eh 
CD 
CJ O 



g 

CD 
Eh 
CJ 



CD CJ 

CD CJ 

Eh fit 

CD O 

CD CJ 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



6 



PCT/GB2003/002804 



^ Eh 

cj a 

u cd 

a cd o 

CD cj 

a CD 

fa Eh <J 

H ft 

CD CJ 



a cd 



Of 



CJ 
CO CD 



H 



CD O 

CD CJ 

CJ CD 

rfl Eh 

ft Eh 

CJ CD 



6 



J*2 



CO CJ U 

Eh rfj 

ft B 

CD CD a 

CD CJ 

Eh ft 

CO CD CJ 
Eh 

CJ CD 

CD CJ 



fa 



CD 



fa 



CD CJ 



6 



6 



CO CD CJ 

< Eh 

CD CJ 

O rij Eh 

CJ CD 

Eh < 

CO CD CJ 



P4 

U CD 

U CD 

CJ CD CJ 
Eh 

Eh ri! 

Eh CJ CD 

ft Eh 

U CD 

Ej < 

rtj eh 



U CD 



CO 



CD O 
CD 
Eh 

CO CD 



a 



EH 

CJ CD 



fa 



fa 



3 



CD CM CJ CD 



•H 
rH 

n 



CJ CD 

CO CJ CD 

El ^ 

cj a 

CO U CD 

Ej g 

red Eh 

A* CJ CD 

CJ CD 

Eh < 

CO CJ CD 

Eh < 

CD CJ 

Oi Eh 

CJ CD 

CJ CD 

B U CD 

tt Eh 

CD CJ 

£h rtj 

*C Eh 

CD CJ 

a ri! Eh 

CJ CD 

U CD 

H Ej ril 

ri; Eh 

CJ CD 

P Eh 

CD CJ 



CO 



H 



CJ CD 
Eh ft 



0 

CJ 



cd a 

P5 CD CJ 

CJ CD 

*C Eh 

« ^ Eh 



H Eh 



CD 



CJ 



cd a 

EH (< 



Pi 



CJ CO 
Eh 

3 CD 



O CD 

EH < a 

ft Eh 

CD CJ 

CD CJ CD CD CJ 

Eh < CD CJ 

CD CJ CJ CD 
fa 




ft Eh 
CJ CD 
CD CJ 

CD CJ 
Eh ft 
CD CJ 
Eh 
CD 



6 



CJ CD 



i 



3 



Eh 

EH 

CD CJ 



Eh CJ CD 

ft Eh 

CD U 

pi CD CJ 

CJ CD 

Eh ft 

0* CJ CD 

CJ CD 

Eh ft 

, CD CJ 

Pi 

CJ CD 

CD CJ 

CD CD CJ 

CD CJ 

CD CJ 

S ^ ^ 
Eh 

CD CJ 



Of 



CJ 



6 



CO 



a 



CD CJ 

a eh 

CJ CD 

ft Eh 

<3 Eh 

O CD 

Eh ft 

CJ CD CJ 

Eh ft 

CJ CD 



CO 



6 



< Eh 
Eh i<! 
U CD 

< Eh 
Eh ^ 
CD CJ 
CJ CD 

< Eh 
Eh f< 
CJ CD 



Q ^ 
CD 
ft 

CD 
Eh 
CJ 



Eh Eh ft 
U ft CJ CD 



Eh 
CJ 



CD CJ 

eh 

CJ >< ft Eh 

Eh ft 

CJ CD 

ft Eh 



CJ H 
<< 
CD 




cd a 

oi cd a 

CJ CD 

ft H 

W Eh 



CD CJ 

CD CD CJ 

CD CJ 

Eh ft 



CD 



CD CJ 



CD CJ 

W H 

*3 E-" 

Eh ft 

hM 

Eh 
O CD 
CO CD CJ 
^ E-J 
CD CJ 



fa 



CJ Eh 



ft 



6 

CJ 



*C EH 

CD CJ 

CD CJ 

ft Eh 

Eh ft 

CD CJ 



O CD 
CD CJ 



CD 



fa 



CO 



CO 



CO 



U CD 
Eh ft 



CD CO 

CD 

CD 

CJ CO 



a 



I 

EH 



Eh 



Of 



CJ CD 
i3 Eh 
CD CJ 



P 



CD 



U 
4J 

» 

Pi 



UO 



^1 



o 



CN 



CO 



CO 



no 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



7 



PCT/GB2003/002804 



E-i 

. H 

U CP 

CJ CD 

CD U 

CD CJ 

a eg 

Eh f< 

EH S 

CD O 

CJ CD 

< H 
Eh rfl 
CJ CD 

< Eh 
Eh *C 
CJ CD 
U CD 
EH fed 
CD CJ 

CJ CD 

Eh ril 

CD CJ 

CJ CD 

CD CJ 

CD CJ 

Eh f*£ 

Eh 3 

CJ CD 

CD CJ 

< Eh 
CJ CD 

Eh 



a cd 

Eh rfj 

CD CJ 
Eh 

U CD 

Eh i=C 

CJ CD 

< Eh 
Eh f£ 
CD a 
CJ CD 
g Eh 
Eh <£ 
CJ CD 

CD CJ 
B 
Eh 
Eh 

Eh , 

< EH 

CD a 
EH 

ss 

Eh fiC 
U CD 
CJ CD 

JSC Eh 

a cd 

CD CJ 

Eh rf3 

CJ CD 

Eh d 



cd a 

CD CJ 

*C Eh 

CJ O 

Eh ril 

CJ CD 



On CD CJ 

CD U 

CJ CD 

< Eh 





Eh 

Eh to 



O CD 

Eh rtj 

ft Eh 

CD CJ 



C« f'i EH 
CJ CD 
CJ CD 



CD CJ 

W ^ Eh 

Eh S 

> B § 

CD CJ 

O CD 

CO CD CJ 

< Eh 

CD a 



CD CD U CD CD CJ 
Eh CD CJ 

CD U U CD 

6 Eh fa 
CD 
CD CJ 

CD CD 
Eh 
CD 
CO rf) 
CJ 




CD CJ 



CD CJ 



CD CD CJ CM < Eh 
CJ CD 
Eh 




■H 



a. 

CJ CD 

CD CJ 

Eh rtj 

CO O CD 

Eh rf 

CD CJ 



Eh <C EH &(, 

CJ CD CJ CD 

CJ CD CJ CD 

Eh < 

cj cd a 

CD CJ 

' Eh 

. Eh 

EH < 

CJ CD 



CO 



CJ CD 

CJ CD 

Eh < 

CJ CD 

CO CJ CD 

Eh < 

< Eh 

CM CJ CD 

CJ CD 

EH < 

CO CJ CD 

Eh < 

CD CJ 



ft CJ 
CJ 
. CJ 
< CD 



c* 



H 



CJ CD 

rf! Eh 

CD U 

6 ' 

CJ 



EH 



Eh 
. CD 
Q Eh 
CD CJ 




Eh 

CX CJ CD 

CJ CD 

< EH 

Eh < 

CD CJ 

CD U 
Eh 
Eh 
CJ 



J CD U 

04 CD U 

CJ CD 

rfj Eh 





a d 

CJ 
CO CD 



Eh < Eh 
CD CO CJ CD 




to 

I 

to 
(d 
ft 



















TT 


CO 


a 








t 






$ 


ot 


J— 5 


Eh 






% 


Eh 






Eh 




CD 


CD 


CJ 












CD 


O 


C3 




EH 

Eh 


CO 


Eh 

S2 


6 












6 


GT 


fa 














CD 


cj 




CD 


u 





fa 



CK < Eh 

CJ CD 
Eh 

CO CD CJ 
Eh 

<3 EH 

CJ CD 

CD CJ 

CD a 

CD CJ 



ft 



CD 



H 



OJ 



fa 



CO 



CO 



CD CO 

CD 

CD 

CJ CO 



H 



to 

9> a 

6 

o 

a h 



Q 



< Eh 
CD CJ 

< B 
CJ CD 
CJ CD 
Eh < 
rf) Eh 
CJ CD 

< H 
CD CJ 



O < Eh 

CJ CD O 

CJ CD 

^ *<! ^ - 

Eh rf EH 

CD CJ 

IS CD O 

Eh CD 

CD O 

<C Eh 



10 



o 

CN 



in 



o 

CO 



o 



10 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



8 



PCT/GB2003/002804 



rf En 

*C Eh 

a cd 

U CD 

cd a 

cd u 

cj o 

eh ^ 

cd a 



CJ CD 

< Eh 
Eh < 

Eh 

. Eh 

Eh < 

U CD 

CJ CD 

CD O 

CD CJ 

CD CJ 

CD CJ 

H < 

CD CJ 
Eh 

SfS 

CD CJ 

CD CJ 

< Eh 
U CD 

" Eh 

EH 

CD 



Eh 
CD 
Eh 
CJ 



6 



EH fit 

a cd 

CD CJ 

CJ CD 

< Eh 

Eh rfj 

CJ CD 

CD CJ 
Eh 
Eh 



rtj Eh 

Eh 

< EH 
CD CJ 

Eh rfj 

U CD 

CJ CD 

< H 

< EH 
CJ CD 
CD CJ 
Eh < 
CJ CD 
Eh < 



cd a 

< Eh 
CJ CD 
CD CJ 

< Eh 
U CD 

< Eh 
CJ CD 
CJ CD 

< Eh 
U CD 
Eh < 
CJ CD 
Eh < 
CJ CD 

< Eh 
CJ CD 
Eh 

EH ^ 



Pi CD CJ 

CD CJ 

CJ CD 

< H 

Eh 

idj Eh 

H CJ 0 

Eh < 

rf± Eh 

2 Eh 

^ EH 

CD U 

CD CJ 

Eh rtj 



fa 



CD CJ 

< Eh 

CJ CD 

ft, Eh Eh 

CD O 

CD CJ 

CD CJ 

Eh (3 



CD CJ 

W CD U 

< Eh 

< Eh 
CJ CD 
CJ CD 
*C Eh 

CD CD CJ 

CD CJ 

CD CJ 



CJ to 
Eh 

3 CD 



O CD < Eh 

Eh < CX < Eh 

Eh CJ CD 

CD O CJ CD 

cd a cd cd a 

Eh < CD CJ 

CD CJ 

< Eh 

CJ CD 

CD CJ 



CD 



fa 



Pi 



CJ CD 

CD CJ 

CD CJ 

Eh fit 



Eh 

CD CJ 



CD U 



CD CD CD 



at 



cj 

o to cj 

6 3 



8 



* 6 

Eh p$ 
CJ CD 



EH rf| 

CD CJ 

CJ CD 

< Eh 

Eh f4 

CJ CD 



fa 



Eh rfj 



CD W 

CD 

CD 

CJ CO 



cd a 



w ft 



H Eh 



CD 



a 

EH 



, CD CJ 

Pi CD CJ 

CJ CD 

ri! Eh 

W d Eh 



fa 



fa 



53 

3 



!*i 



CD CD CJ 
CD U 



CD 



CO 



fa 



Eh O CD 

(dj Eh 

CD CJ 

Pi CD CJ 

Eh < 

Cm O CD 

U CD 

CD CJ 

CD CJ 



< Eh 

CO CJ CD 

H < 

ftf EH 

cm CJ CD 



CD CD CJ 

CD CJ 

Eh rfj 

CO CD CJ 

*C Eh 



Eh 



CD CJ 



CJ pi CD U 
CJ CD 
Eh rtj 



CJ CD 



CJ H Eh rtj 
CD 



(< Eh 
CD CJ 



CM 



a 



< EH 

Eh f< 

CJ CD 

< Eh 
Eh rf 
CD CJ 
CJ CD 



O CD 



fa 



fa 



Eh rf! 

Cm CJ CD 

CJ CD 

f£ Eh 

3 Eh 

O CD 

CD CJ 

Eh rfj 

U CD 

Eh < 



Eh 



6 



3 



CJ CD 
Eh 
<2 Eh 
*2 Eh 
CD CJ 
Oi < Eh 
CJ CD 
CD CJ 



CD CJ 

Pi CD a 

CJ CD 

< Eh 

W Eh 



H Eh 



fa 



fa 



O 
iH 

g 

o 

s 



a) 
w 




a) ci 

o o 

cJ q 

CD 0) 

&* 

Q> CD 

ra g. 

d xi 

cd -u 



3 



6 



CD 



Oi 



fa 



Pi 



CM 



u 



Cm 



in 
i 



SU pi 



in 



o 



in 



o 

CO 



in 



SUBSTITUTE SHEET (RULE 26) 



o 



WO 2004/003019 



9 



PCT/GB2003/002804 



a cd 

CD o 

CD O 

CD U 



H fit 



cj 



CD CJ 

H < 

fit Eh 

U CD 



Pi 

CD U 

CJ CD 

^ cd y 

Eh < 

CJ CD 

C/3 CJ CD 

CD CJ 

fit Eh 

CJ CD 

% < Eh 

< Eh 

JS CD CJ 

fit Eh 

Of En 



CD CJ 
CJ CD 



CJ CD 



CJ CD 

Eh CJ CD 

f=3j Eh 

fit Eh 

CD CD CJ 

CD CJ 

CD CJ 

0( fit Eh 

CJ CD 

O CD 



CD CJ 
CD CJ 



6 



CD CJ 

CJ CD 

CJ CD 

Eh rfl 

CD CJ 

CD CJ 

CD CJ 





CD CJ CJ CD 
CJ CD fit CD CJ 
Eh < 



CJ CD 

Eh »< „ 

CJ CD CJ 

^ Eh 

CD CJ 

Eh >h 

CJ CD 

CD CJ 



CD CJ 

Eh fit 

U CD 

fit Eh 
Eh 



Eh < 

£ g 

< Eh 

CD CD CJ 

CD CJ 

CJ CD 

to CJ CD 

Eh fit 

CJ CD 

CJ CD 

CD CJ 

fit Eh 

CJ CD 

CD CJ 



CJ CD 

>h rfl Eh 

Eh rf 

Eh fit 

CD CD O 

CD O 
Eh 

, « 

CD CJ 



cj 



Eh fit 

CD U 

Eh rtj 

CJ CD 

to CJ CD 

Eh rfj 



Fki CD 
CJ 
CD 

CD CD 
CD 
Eh 

CD CD 
CD 



N EH <: 

< Eh 

> CD 8 

CD U 

CJ CD 

tfj CD CJ 

CJ CD 

CJ CD 

Eh fit Eh 

O CD 
Eh 

CJ Q CD CJ 



CJ 
CD 



CJ 
CJ 



CD CJ 
fit Eh 



fit H CD CJ 



CJ 
CJ 



P4 



CJ CD 

Eh fit 

CJ CD 

Eh fit 

CD U 

CJ CD 

CD CJ 

Eh fit 

CJ CD 

CJ CD 



W U CD 

Eh fit 

CD CJ 

CD CD CJ 

CD CJ 

CD a 

CD CD CJ 

CD CJ 

Eh < 

CJ CD 

Pi 

CJ CD 

CD CJ 

Oi *C Eh 

CJ CD 

w < Eh 
> Eh 

CD U 

CD CJ 



CJ CD 

CJ CD 

fit O V 

Eh rfj 

CD CJ 

fit Pi CJ CD 



CD 



J Eh fit Eh rf 
^ £h 2| CD CJ 



U 



•rl 
H 

CO 



CJ CD 

CD CD CJ 

CD CJ 

fit Eh 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD a 

Eh fid 



Eh fit CD CJ 

>-q CJ CD to CJ CD 

CJ CD Eh cC 

CD CJ CJ CD 

to fit Eh t> Eh rij 

CJ CD CD CJ 

< Eh CJ CD 

£ fit Eh Eh CJ CD 

CD CJ fit Eh 

CJ CD 

> Eh rf 

CD CJ 

CD CJ 

h3 Eh < 

CJ CD 

CJ CD 

CJ CD 
Eh , 

fit Eh 

fit, H 

CD CD CJ 

CD U 

CD CJ 

Oi *3 Eh 

CJ CD 

O CD 

CD CD CJ 

CD CJ 

CD O 

13 CD CJ 

Eh < 

V CD 

^ < Ej 

Eh rf3 

CJ CD 

Pi fit fr* 

CD O 

Eh < 



Pn CJ CD 

U CD 

CJ CD 

to eh f=U 

CJ CD 

Eh rij 

CJ H fit Eh 

a cj o 



fit Eh 
CJ CD 





CD 

cj to 



CD CJ rf) 

CJ CD 

O CD 

Eh fit O 

CJ CD 

fit Eh 

CD CJ 

f< Eh 

CJ CD 

CD CJ 

CJ CD 

£C EH 

Eh fit 

CD O 

cd a 

Eh 
Eh 

CJ CD 

fit EH 

CD CJ 



CJ CD 

CD CJ 

Eh < 

CD CJ 

Eh fit 

CJ CD 

fit Eh 

EH fit 

Eh icq 

rfj Eh 

E-J 

< E-J 

EH I< 

CD O 

CD O 

cj a 

CD CJ 

U CD 

CJ CD 

fit Eh 

CJ CD 

fit Eh 

CD CJ 

CD CJ 

rfj Eh 

CD CJ 

CJ CD 

CJ CD 

CD CJ 

Eh fit, 

CD O 

CJ CD 

CD O 

F fit 

CJ CD 

CJ CD 

CD U 

fit Eh 

CJ CD 

fit Eh 




cd a 

to CJ CD 

Eh fit, 

CJ CD 

EH fit 

CD CJ 

CJ CD 

CJ CD 

fit Eh 

CJ CD 

Eh fit 

CD U 

CD CJ 

U CD 

Eh O O 
Eh 

. Eh 

CD CD a 

CD CJ 

. C2 CJ 

O? fid EH 
CJ CD 
O CD 



$ 

u 

P4 



CD 



u a <t b 

CD O CD 

CD fit Eh 

< Eh CJ CD 

CD fit Eh 

CD CD CJ 



W 
CD 
CD 

& 

> 

. — » 
U 

■a u 

-rl 

H CD 
& 

n CD 



CO 
A H 



U CD 

Eh *3 

CD O 

CD U 

CD CJ 

CD CJ 

CD CJ 

CD U 

Eh f< 

O CD 

U CD 

CD CJ 

fit Eh 

CJ CD 

< Eh 
Eh<< 

CD O 

CD CJ 
Eh 
Eh 

U CD 

CD CJ 

CD O 

fit Eh 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

B fit, 

CJ CD 

Eh < 

CD O 

fit Eh 

CD CJ 

CD CJ 

Eh tfj 
Eh 



CD U 

is CD CJ 

Eh fit 

& ^ 

to CJ O 

H PC 

O CD 

> Eh 

CD O 

CD CJ 

CD CJ 

W ri! H 

CD O 

fit Eh 

J Eh rfJ 

U CD 

Eh fit 

CD O 

CD 

CD O 

CD CJ 

*2 Eh 

CD O 

CD CD CJ 

CD O 

Pi CJ CD 

CJ CD 

Eh fit 

fit CJ CD 

CD CJ 

CD O 

O < Eh 

U CD 

CJ CD 

« CD CJ 

88 




s 

4J 

u 

At 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



10 



PCT/GB2003/002804 




d CJ CD 

CD CJ 

CJ CD a 

E-» *C 

O CD 

< E-J 
Eh 

. 2 



EH 



a. 

CD CJ 

rij CJ CD 

CD CJ 

CJ CD 

H U CD 

<: Eh 

CJ CD 
Q < Eh 

o a 
cd a 

fa < Eh 

cd a 

, U CD 

O CD 

U CJ 

Eh rfj 



CD 



U W 



CD 



fa 



U CD 

cd a 

Eh < 

CD CJ 

Eh O 

CJ CD 
Eh 

Eh < 
EH 

< Eh 



CJ CO U 
Eh Eh 
CD CD 

CJ O* 



< CD CD 



CD 



Q 



Pj 



CD < CJ 



CD CD CD 



CD CD 

< a < 

o CJ 
CJ 

O CO u 
U Eh 

U , CJ 

y > ^ 

< CD 

CD CD 



CD fa rf{ 



CD CJ 

U CD 

Eh f< 

CJ CD 

§6 

CJ CD 



CD 



u cd a 
cp a 
cj cd 

6§ 



* 

CD 
CD 
CD 

S 



CD 



CD 
CD CD 



CJ CD 
CJ CD CD 



s 



e£ Eh 

^ B >H 

CJ CD 

U CD 
Eh p 

Eh < 

ril Eh 

is* Eh fa 

u a 

ril Eh 

CD CJ fa 

CJ CD 

C5 CJ , 

U C5 >h 

CJ CD 

CJ CD 
Eh 



CD U 

CJ CD 

Eh j< 

CJ CD 

f3 Eh 

CD CJ 

Eh rij 



CD CJ 

is o y 

Eh fdj Eh 

CD CJ 

CD CD CJ 

CD CJ fa 

CD CJ 
Eh 



CJ CD 

Eh < 

icC Eh 

CJ CD 

CJ CD 

*C Eh 



fa 



CD 



fa" 



a) 

s 

1H 

cd 
P. 




CD CJ 

f< Eh 

CJ CD 

CD O 
Eh 

CJ CD CJ 

Eh < 

U CD 

CO CJ CD 

eh rf: 

CJ CD 
Eh 

CJ CD 

Eh < 

Ph CD CJ 

U CD 

CD CJ 
Eh 



^ <| Eh 

Eh < 

CD CJ 

m CD CJ 

CJ CD 

Eh < 

CD CD O 

CD CJ 

eh <: 

CJ CD 




CD CJ 

CO CJ CD 

Eh < 

CJ CD 

Eh rfl 

CD CJ 

CJ CD 

Eh CJ CD 

r=C EH 

CJ CD 

Eh < 

CD CJ 

CD CJ 

Eh < 

CJ CD 

CJ CD 

CJ CD 

Eh 

, EH 

CD CD CJ 

CD CJ 

CD CJ 

Oi rfj Eh 

CJ CD 

O CD 

CD CD CJ 

CD CJ 




CJ CD 

i* < Eh 

Eh f=C 

a cd 



CJ 
CO CJ 

8 

A 

CD 
u CD 

EH 

a 

CO CJ 
Eh 

U 

J EH 

CJ 
Eh 

*g 
CD 

HH" EH 
CJ 

a 



CJ >H rfj 



CD 

CD 
CD 



Eh 



En CJ CD 
*C Eh 
CJ CD 



a co 

Eh 
CD 

CJ CD 

6 

t=C CD 
CD 

S2 

CO 

CD CD 

< 

CJ 

CD CO 

a 

CD H 
CD 



CD CJ 

< Eh 

Eh < 

CD CJ 

CD CJ 

Eh p< 

CD CJ 

CD a 

EH isC 

CD CJ 

rfj Eh 

Eh *S 

CD CJ 

CD CJ 

Eh < 

CD O 

e< Eh 
Eh 

a 



CO 
CD 
CD 



u CD 

Eh rtj 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

EH rtj 

a cd 

a cd 

CD CJ 

*C Eh 

U CD 

Eh- rf! 

CD CJ 

CD CJ 



CO 



■a 

•H 
H 



^ CJ CD 

CD CJ 

6Eh 
CD 

EH rtl 

U CD 

> ir* < 

CD CJ 

CD CJ 

£ a CJ 

Eh < 

CD CJ 

fa < Eh 

CD CJ 

< Eh 

^ Eh < 

CJ CD 



CD 



CD 



rn CD 



CO 



m 
i 

A 

3 

•rl 



fa 







CD 






CJ 


CD 


CD 


U 


CD 


U 




CD 


CJ 


55 


U 




EH 


•3 


Eh 






Eh 


CD 


U 




O 


CJ 


CD 


CJ 


CD 


CD 


CJ 


CD 


a 




S3 


CJ 


CD 


CJ 




6 


Eh 


CD 


a 


Pi 




CD 


Eh 


< 




u 


CD 


O 






H 


< 




< 


U 


CD 


1 


o 




CD 


U 




Eh 




CD 


CJ 


CD 


CJ 


a ^ 


Eh 


CD 


a 




O 


CD 






Pi 


a 


CD 




| 


CD 


CJ 


CD 






u 


CD 


EH 






CJ 


CD 


a 


CD 


> 


Eh 




CD 


U 




CD 


a 




EH 




CD 


CJ 


cj 


CD 




CD 


CJ 


CD 






Eh 




Eh 






Eh 




CD 




fa 


Eh 




CD 


CJ 




Eh 




< 


EH 




CD 




CD 


U 




EH 


< 



t— i ^— < cN cN rn m ^ 

SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



11 



PCT/GB2003/002804 



cd cj 

rf En 

^ EH 

CD O 

CJ CD 

« £h 



CJ CD 

Q Eh 

CD CJ 

CD CJ 

fa < H 

CD CJ 

, U CD 

< o cd 

o y 

eh *c 



CD CJ 

CJ CD 

Pi CD CJ 

CJ CD 

CJ CD 

CO CJ CD 

Eh tfl 



CD CJ 

CO O CD 

EH < 

CJ CD 
^ Eh 

CD CJ 

CJ CD 

Eh CJ CD 

*C Eh 

CJ CD 

> Eh rtj 

CD CJ 

CD CJ 

h5 Eh < 

CJ CD 

CJ CD 

EH CJ CD 
Eh 

< Eh 

CD CD CJ 

CD CJ 

CD CJ 

O Eh 

CJ CD 

a cd 

CD CD CJ 

CD a 

CD CJ 

CD CJ 

Eh <| 

CJ CD 

3 



fa 




Q 



CD CJ 

CJ CD 

CJ CD 

CD CJ 

CD O 

CD CJ 



fa 



a cd 

rfj Eh 

CD U 

rtl EH 

CJ CD 

CD CJ 



< CJ CD 

CD CJ 
Eh 

CJ CD CJ 



U CD 
r* < H 



rt3 Eh 



CD 



CJ o 

P Eh 

CD CJ 

CD U 

W rtj Eh 

CD CJ 

CJ CD 

rtj CJ CD 

CD CJ 

Eh < 

Pi CD CJ 

CJ CD 

CD CJ 

^ Eh <; 

CJ CD 

CJ CD 

CO CD CJ 

f3 Eh 

3g 



CD 
CD CD 



O CJ 



53 



< Eh 
CD CJ 
Eh < 



a 



EH rfj 

O CD 

Eh i< 

CD CJ 

CD CJ 

CD CJ 

rtj Eh 

CD U 



U CD 
Eh rtj 
CD U 



a 



2 



CD U 

Eh t$ 

CJ CD 

CD CJ 

CJ CD 

ss 

EH 
rf| Eh 



f£ Eh 

p£ Eh 

CD CD CJ 

CD CJ 

CD CJ 

O? fcC Eh 

cj CD 

CJ CD 



CD CD CJ 



CJ 



CD . Eh rtj 

rtj CJ CD 

CJ CD CJ 

Eh CD CJ 

CJ Ot Eh 

CJ CJ CD 

ril CJ CD 

2 « C5 u 

CJ CJ CD 

*3 U CD 



CJ CD 

Q < Eh 

CD CJ 

CJ CD 

Ph CD CJ 

CJ CD 

CJ CD 

CO CJ CD 

Eh < 



fa 



fa 



f3 EH 

CD CJ 

PS CD CJ 

< Eh 

CD CJ 

« ^ Eh 

Pi CD S 

CJ CD 



Eh « CD 
U CJ 
Eh CJ 

E-] CD CD 
CD 

Eh . CD 



9 



3 



1 



CD CD 



CJ CD 

D < Eh 

CD CJ 

CD CJ 

fa rfj Eh 

CD CJ 

CJ CD 

rfj CJ CD 

CD CJ 

Eh < 



CD Pi CD 



CD CD CD 



CD H E-i 



CD 



< EH 



CD CJ 
CJ CD 



CJ CD 
CJ CD 



CJ CD 
< Eh 



CD CJ 

CO CJ CD 

Eh < 

CJ CD 

> Eh < 

CD CJ 

CJ CD 



CD O 
cj < 

CJ CO CJ 



CD fa f< 

Eh CD 

cj u 

3 CD 
CD 



°6 



Eh f< 
CJ CD 



Eh j< 

CJ CD 

CD CJ 

CJ CD 



Eh 



CJ CD 
CJ CD 
CJ CD 



s 



CD 



a 



CD 
fa rfj 



CD > Eh 

cj CD 

Eh CD 

CD & CD 

CJ Eh 

S CD CD 

a CD 

Eh CD 

CJ Eh 



CJ 

U fa Eh 



Eh 

*C Eh 

CJ CD 

CJ CD 
Eh 

Eh g 

< Eh 

< Eh 
CJ CD 

< Eh 
CD CJ 
CJ CD 
CD CJ 
CJ CD 
CJ CD 
CJ CD 
Eh st; 

CJ CD 

Eh < 

< Eh 
CJ CD 
CJ CD 

< Eh 
CJ CD 



Cm 



Pi CD CJ 

CJ CD 

CJ CD 

CD CD CJ 

cd a 

CD CJ 



fci 



fa 



Eh 
. Eh 
CD CJ 
Eh & 



fa Eh fit 

Eh g 

< Eh 

CD CD U 

CD CJ 

U CD 

CO CJ CD 

Eh »sC 

CJ CD 

4 CJ CD 

q cj 

rtj Eh 

^ U CD 

CD CJ 

Eh rtj 

CJ CD O 

Eh < 

CJ CD 

CJ CD 

Eh < 



CO 



CD U 

CJ CD 

CO CJ CD 

EH < 

a cd 

Q tfj H 

CD U 

< Eh 
< CJ CD 

CD CJ 

CJ CD 

>h Eh 

Eh < 

CJ CD 

>h t< Eh 

Eh ^ 
Eh 

Eh U CD 

O CD 

CO CD CJ 

< Eh 
Eh i< 

CD CD a 

CD CJ 

Eh < 

CD CD CJ 

CD CJ 

Eh < 

CD U 



U CD 
^ H 

CJ CD 

Pi CD CJ 

U CD 

CD CJ 

J E-J 

U CD 

U CD 



CO 

< Eh 
Eh 

CD CD CJ 

CD CJ 
Eh 

CO CD CJ 

< Eh 
Eh i< 

H H 

< £h 

CD Eh rtl 

< rfj CJ CD 

CJ CD CJ 

CJ < £h 

CJ CO CJ CD 



CD Eh < 

< rtj CJ CD 

CJ CD CJ 

EH CD CJ 

CJ C* < Eh 

U U CD 

CJ CD 

Pi CD CJ 

a a cd 

CJ CD 



O CO CJ CD 

O Eh < 

Eh CD CJ 

CJ Eh < 



V4 

18 



U 

s 

4J 
(8 

P4 



a) 

5 

Pi 



in 



in 



o 

CN 



in 



SUBSTITUTE SHEET (RULE 26) 



o 



WO 2004/003019 



12 



PCT/GB2003/002804 





CD 


CJ 








fit 






CJ 


CD 






CD 


CJ 




s 


Eh 


















Eh 






Eh 








Eh 






CD 


CJ 




rfj CJ 


CD 






rh 


CJ 






Bh 






cj 


CD 


3 






c_i 








r i 


TG 






s 






tH 








r i 

s 


a 








&H 










r ■ 


a 












> 










CD 


n 




fit 




u 




r i 
LJ 


rh 

CJ 






r i 

CJ 






r \ 


rh 

CJ 




fcH 




rn 
LU 








r i 






r ) 


rh 




[—1 




r i 
tH 






rh 


r 1 






rh 


r i 




fVl 


a 










r i 






r i 






trf 1 


r j 


CD 






rh 


CJ 






fi 


erf 
fi** 




(V 
r-M 


rh 


r i 






r i 


rh 






CD 


r i 






EH 










CD 








rh 
CJ 






rh 








s 


L. » 








CD 


r* 




fi*1 


Eh 




B 










EH 






r l"i 


CJ 


C 1 




Ph 










Eh 








Eh 


> 


Oi 




Eh 








CD 






CD 


CJ 




1-3 


Eh 


3 






CJ 










1 


Fh 

1* ■ 










Eh 








CD 


3 


CD 




Eh 






CJ 








CD 


CJ 


a 


EH 


CJ 


CD 






< 


EH 


CD 




CJ 


CD 






Eh 






cf 


Eh 








CJ 


IS 




EH 






r 3 


EH 






u 


CD 


1* 


CO 


CJ 








Eh 






Eh 




Q 




< 
















fit 


Eh 






CJ 


CD 




Q 


^ 


Eh 






CD 


CJ 




Pi 


CJ 


CD 


> 


CD 


a 






CJ 


CD 






CJ 


CD 


CO 


CJ 


CD 






Eh 


fit 






CJ 






H 


H 


$ 






6 






Eh 




CD 


CJ 


CD 








Eh 




fa 






Eh 


Eh 










P4 









CD CJ 

a cd 

Eh < 

CJ CD 
Eh 

cd a 

U CD 

CJ CD 

' Eh 
CD 



6 



Eh 
CD 

CD CJ 

Eh < 

cj CD 

CJ CD 

CJ CD 



3 



Eh 
Eh 

CD CJ 

cd a 

CD CJ 

< Eh 

a cd 

a cd 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

Eh < 

CJ CD 

< E-J 

EH fit 

CJ CD 

fit Eh 

CD a 

Eh fit 

Eh tf| 

Eh fit 

CD CJ 

Eh f<C 

CD CJ 

cd a 

< Eh 
fit Eh 



Eh 

CD CJ 

fit Eh 

§ £ 

Eh fit 

CJ CD 

*C Eh 

CD CJ 

CD CJ 

< Eh 




CJ CD 

Q < H 

O U 

CD CJ 

W rij Eh 

CD CJ 

CJ CD 



CJ 
»-q Eh 
CJ 

& CD 
CJ 
^ CD 

h1 Eh 
CJ 

a 

W CJ 
Eh 
CD 

CD CD 

a 

CD 
CD CD 
CD 
Eh 

CJ 



CD P5 CD 

a cd 

CD CD CD 



CJ 
< 
CD H 
CD 



CD 
Eh 



CD CD 

f=U rfj cj 

CJ CD 
CJ 

CJ CO CJ 

CJ Eh 

CJ CJ 

O > Eh 

< CD 

CD CD 




CJ CJ 

CJ Eh 
<C >* < 



s 



rf O CJ 

CD W O CD 

CD Eh f3 

CJ CJ CD 

Eh > Eh e$ 

CD CD CJ 

Eh CJ CD 

Eh Eh CJ CD 

CJ nC Eh 

f< U CD 
Eh > Eh 

Eh CD CJ 

Eh CD CJ 

CD J Eh < 

U CD 

CJ CD 

CJ CD 

Eh 

< Eh 
O CJ 
CD U 
CD CJ 

< Eh 

a cd 

a cd 

CD CJ 

cd a 



Eh 



Eh ft 

Eh < 

cd a 

(dj CJ CD 

CD CJ 

A Eh ^ 

^ cj cd 

CD U 

Eh < 

CJ CD 



B 



u 
o 
d 

Cm 




fa EH 

EJ g 

EH 

CD CD CJ 

CD CJ 

CJ CD 

CO CJ CD 

Eh f< 

CJ CD 

CJ CD 

CD CJ 

islj CJ CD 

CD U 
Eh 

CJ CD CJ 

Eh (3 

CJ CD 

to CJ CD 

Eh < 



W < Eh 

CJ CD 

CD CJ 

Ct! CD CJ 

^ 

Eh < 

CD CD CJ 

CD CJ 

CD CJ 

CD CJ 



CJ CD 

Q & Eh 

CD a 

CD CJ 

W < Eh 

CD U 

, CJ CD 

<C CJ CD 

CD CJ 

Eh < 

Ph CD CJ 

U CD 

CD O 

Eh *< 

CJ CD 

CJ CD 

CO CD CJ 

t< Eh 

CJ CD 

fit Eh 




Eh 
Eh 
Eh 

CO CJ CD 

Eh fiC 
Eh 

Pi CD U 

CJ CD 
CJ 
CJ 

CD CJ 

CD CJ 

Pi CD CJ 

CJ CD 

EH < 

CD CD CJ 

CD CJ 

CD CJ 

P< CJ CD 

CJ CD 



in 



s 












Eh 
CJ 


Pi 




8 


a 






CJ 




CD 










fH 




1 










Pi 


CD 






CD 
CJ 

Eh 


CJ 
CD 


> 


Pi 


CD 


6 






CJ 


CD 


CO 



CD CJ 

CJ CD 

CJ CD 

CD U 

CD CJ 

CD CJ 

rf Eh 
Eh 

CD CJ 

Eh < 

CD CJ 

CJ CD 

O CD 

Eh rtj 

a cd 




h 
a) 

CJ 
•H 
H 



CO CJ 

CD CD 
CD 
CD 

CD CD 
CD 
Eh 
CJ 

Pi 

CJ 
CD 

a < 

CJ 

g 

CD 
Eh 
Eh 



CD Eh 

<C CD CD CJ 

CJ CD CJ 

U fdj Eh 

CJ CO CJ CD 



a 
a 
a 
< 

CD 

CD rS 

CJ 
Eh 

cd ra 

Eh 



6 

y 
3 



U 
CD CD 
CD 
< 
CD CD 
CD 
CD 
CD CD 
CD 
Eh 

CO CJ 
EH 
C9 

H 

CD 
, CD 
>4 Eh, 
Eh 
CD 

* B 

CJ 
CD 

a < 

CJ 
CD 

> ? 

CD 
CD 

w pi: 

CD 



CD 



Eh rfl 

CJ CD 

Eh < 

CD CJ 

CD CJ 

CD CJ 

Eh rtj 

CD CJ 

< EH 
CD U 

< Eh 
Eh < 
CJ CD 
Eh < 
CD CJ 



CD 

a 
a >4 

Eh 
CJ 

U CD 

u 

CJ 

a Cm 



CD CJ 

CD CJ 

fit Eh 

fMl H 

CD O 

CD CJ 

CD CJ 

■3 Eh 

CJ CD 

CJ CD 



CD Eh 

<d fit CJ CD 

CJ CD CJ 

Eh CD CJ 

U Of fit Eh 

CJ CJ CD 

fit , CJ CD 

pj CD CJ 

CJ CJ CD 

rij CJ CD 



CD 
CJ 
Eh 
CD 

3 _ - 

CJ Pi CD O 



Eh <: 

CD CJ 

CD CJ 

CD CJ 

Eh fit 

CD CJ 



CJ 
Eh 
CJ 



fit Eh 
CD CJ 
Eh fit 



t-ht— (CNcNmcoxf"^j- 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

13 



cd cj 

W < E-s 

CD q 

CD CD CJ 

CD CJ 




rij CJ CD 

CD CJ 

Eh < 

CJ CD CJ 

Eh < 

O CD 

>h H 
Eh 

a 



>H 



s 

Eh 



CD CJ 

CD CJ 

rtj CJ CD 

CD CJ 

CJ CD 

Eh CJ CD 

<J Eh 

O CD 

O < Eh 

CD U 

CD CJ 

H < Eh 

CD CJ 

CJ CD 

<; cj cd 

CD U 

Eh f3 




CO 



Eh < 

< g 
Eh rf: 
CD CJ 
Eh rtl 
CJ CD 
CD CJ 
CJ CD 

< Eh 
CJ CD 

En 

f3 B 

CD CJ 

< Eh 

< Eh 
CJ CD 

CO CJ CD 
Eh 

H g 

rfj Eh 

S _ 

i< Eh 

CJ CD 

Q < Eh 

CD CJ 

CJ CD 

PS CD CJ 

CJ CD 

CJ CD 

CO CJ CD 

EH J< 



Eh 



CD CJ 

CJ CD 

Eh < 

CJ CD 

Eh a; 

CD CJ 

CJ CD 

U CD 

< Eh 

O CD 

H < 

CD U 

CD CJ 

Eh < 

CJ CD 

CJ CD 

U CD 



EH 
Eh 

CD CD CJ 
CD CJ 
CD CJ 
a < Eh 
CJ CD 
CJ CD 
CJ 
CJ 



° g 



CO 



CD CJ 

cd a 

Eh rfj 

CJ CD 

< Eh 
Eh *S 
CJ CD 

< Eh 
CD U 
Eh rij 

Eh 

EH (3 

CD CJ 

U CD 

CD CJ 

Eh f< 

U CD 
Eh 




CD 



U Eh 

CD W rf! Eh 

CD CJ CD 

CJ CD CJ 

CJ On CD CJ 

CJ CJ CD 

Eh < Eh 

Eh fc^ flj Eh 



0 



CJ CD 

^ Eh l< 

a cd 

Eh 

Pi CD CJ 

CJ CD 

_ CD CJ 

J Eh <! 

U CD 

U CD 



CD H < 




CJ paj CJ CD 

CD CD U 

CD Eh 4 

< CJ CD CJ 
CD H < 
Eh CJ CD 
CJ >h < Eh 
Eh Eh 

CD Eh f3J 

U >H 

CD Eh f3J 

Eh K Eh 

< > Eh 

O CD CJ 

Eh CD CJ 

rtj < CJ CD 

Eh CD a 

cd a cd 

Eh Eh CJ CD 

CD < Eh 

CJ CJ CD 

Eh Q < EH 

«3 CD CJ 

CJ CD U 

U H <I Eh 

< CD CJ 
CJ CJ CD 
CJ < CJ CD 

CD CJ 

Eh f< 



CJ CD 
CJ CD CD 



CD 



CO 



H 





CD 


CJ 


CO 


CJ 


S3 




EH 






CJ 




> 


Eh 






CD 






CJ 


CD 


EH 


CJ 


CD 




6 


EH 






CD 


> 


Eh 


< 




CD 


cj 




CD 


u 


hH" 


Eh 


«3 




CJ 


CD 




CJ 


CD 




CJ 


CD 


EH 










Eh 




cl 


Eh 


CD 




CJ 




CD 


U 




CD 


cj 


Ot 


fi 


Eh 






CD 




U 


CD 


CD 


CD 


a 


CD 


CJ 




CD 


a 


IS 


CD 






Eh 


H 




O 


CD 






& 












CD 


n 




Eh 




CD 


CJ 




Eh 






Eh 






Eh 


g 




CD 




a < 


Eh 




CJ 


CD 




CD 


CJ 




CD 


CJ 




< 


|H 




Eh 


<=£ 




CJ 


CD 




CD 


CJ 




CD 


CJ 


w 


< 


EH 




CD 


CJ 




Eh 


*s 


CO 


CJ 


CD 




Eh 






Eh 




CO 








< 


Eh 



u 
a) 
a 

u 
n( 

Pi 



w~j o »0 O 



SUBSTITUTE SHEET (RULE 26) 



(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property 
Organization 

International Bureau 

(43) International Publication Date 
8 January 2004 (08.01.2004) 





PCT 



(10) International Publication Number 

WO 2004/003019 A3 



(51) International Patent Classification 7 : C07K 16/24, 

16/18, 16/28, A61K 39/395, C07K 16/46, C12N 15/63, 
15/62, 15/13 

(21) International Application Number: 

PCT/GB2003/002804 

(22) International Filing Date: 30 June 2003 (30.06.2003) 
(25) Filing Language: English 



(74) Agents: MASCHIO, Antonio et aL; D Young & Co, 21 
New Fetter Lane, London EC4A IDA (GB). 

(81) Designated States (national): AE, AG, AL, AM, AT, AU, 
AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CO, CR, CU, 
CZ, DE, DK, DM, DZ, EC, EE, ES, K, GB, GD, GE, GH, 
GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, 
LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, 
MX, MZ, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, 
SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, 
UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. 



(26) Publication Language: 



English 



(30) Priority Data: 

PCT/GB02/03014 28 June 2002 (28.06.2002) GB 

0230202.4 27 December 2002 (27.12.2002) GB 

(71) Applicant (for all designated States except US): DOMAN- 
TIS LIMITED [GB/GB]; 315 Cambridge Science Park, 
Cambridge, Cambridgeshire CB4 0WG (GB). 

(72) Inventors; and 

(75) Inventors/Applicants (for US only): WINTER, Greg 
[GB/GB]; MRC Laboratory of Molecular Biology, Hills 
Road, Cambridge CB2 2QH (GB). TOMLINSON, Ian 
[GB/GB]; Domantis Limited, Granta Park, Abington, 
Cambridge CB1 6GS (GB). IGNATOVICH, Olga 
[BY/GB]; Domantis Limited, Granta Park, Abington, 
Cambridge CB1 6GS (GB). HOLT, Lucy [GB/GB]; 
Domantis Limited, Granta Park, Abington, Cambridge 
CB1 6GS (GB). DE ANGELIS, Elena [IT/GB]; Do- 
mantis Limited, Granta Park, Abington, Cambridge CB1 
6GS (GB). JONES, Phil [GB/GB]; 15 Impington Lane, 
Impington, Cambridge CB4 9LT (GB). 



(84) Designated States (regional): ARIPO patent (GH, GM, 
KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZM, ZW), 
Eurasian patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), 
European patent (AT, BE, BG, CH, CY, CZ, DE, DK, EE, 
ES, FI, FR, GB, GR, HU, IE, IT, LU, MC, NL, PT, RO, 
SE, SI, SK, TR), OAPI patent (BF, BJ, CF, CG, CI, CM, 
GA, GN, GQ, GW, ML, MR, NE, SN, TD, TG). 

Declaration under Rule 4.17: 

— of inventorship (Rule 4.17 (iv)) for US o nly 

Published: 

— with international search report 

— before the expiration of the time limit for amending the 
claims and to be republished in the event of receipt of 
amendments 

(88) Date of publication of the international search report: 

10 September 2004 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



< 
as 



fs| (54) Title: IMMUNOGLOBIN SINGLE VARIANT ANTIGEN-BINDING DOMAINS AND DUAL-SPECIFIC CONSTRUCTS 



O 



(57) Abstract: The invention provides a dual-specific ligand comprising a first immunoglobulin variable domain having a first bind- 
ing specificity and a complementary or non-complementary immunoglobulin variable domain having a second binding specificity. 



INTERNATIONAL SEARCH REPORT 



lnt< a! Application No 

P( 5 03/02804 



A. CLASSIFICATION OF SUBJECT MATTER , 

IPC 7 C07K16/24 C07K16/18 C07K16/28 A61K39/395 C07K16/46 
C12N15/63 C12N15/62 C12N15/13 

According to International Patent Classification (IPC) or to both national classification and IPC 

B. FIELDS SEARCHED 



Minimum documentation searched (classification system followed by classification symbols) 

IPC 7 C07K 



Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched 
Electronic data base consulted during the international search (name of data base and, where practical, search terms used) 

EPO-Internal , BIOSIS, HP I Data, PAJ, MEDLINE, EMBASE 



C. DOCUMENTS CONSIDERED TO BE RELEVANT 



Category 



Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



A 



X 



ELS CONRATH K ET AL: "Camel single-domain 
antibodies as modular building units in 
bi specific and bivalent antibody 
construct s " 

JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN 
SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, 
MD, US, 

vol. 276, no. 10, 

9 March 2001 (2001-03-09), pages 

7346-7350, XP002248402 

ISSN: 0021-9258 

cited 1n the application 

page 7350 

page 7349 



1-21, 
47-56 , 
60,111, 
112 



113-117 

24-26, 

38,40, 
47-56, 
64-67 , 
70-72, 



)( Further documents are listed in the continuation of box C. 



El 



Patent family members are listed in annex. 



° Special categories of cited documents : 

•A" document defining the general state of the art which is not 
considered to be of particular relevance 

*E' earlier document but published on or after the international 
filing date 

*L" document which may throw doubts on priority claim(s) or 
which is cited to establish the publication date of another 
citation or other special reason (as specified) 

■O" document referring to an oral disclosure, use, exhibition or 
other means 

"P" document published prior to the international filing date but 
later than the priority date claimed 



"T J iater document published after the international filing date 
or priority date and not in conflict with the application but 
cited to understand the principle or theory underlying the 
invention 

'X' document of particular relevance; the claimed invention 
cannot be considered novel or cannot be considered to 
involve an inventive step when the document is taken alone 

"Y 1 document of particular relevance; the claimed invention 

cannot be considered to involve an inventive step when the 
document is combined with one or more other such docu- 
ments, such combination being obvious to a person skilled 
in the art. 

document member of the same patent family 



Date of the actual completion of the international search 

8 June 2004 


Date of mailing of the international search report j 

08. 07. 2004 


Name and mailing address of the ISA 

European Patent Office, P.B. 5818 Patentlaan 2 
NL - 2280 HV Rijswijk 
Tel. (+31 -70) 340-2040, Tx, 31 651 epo nJ, 
Fax: (+31-70) 340-3016 


Authorized officer 

Wagner , R 



Form PCT/ISA/210 (second shaet) (January 2004) 



naoe 1 of 5 



INTERNATIONAL SEARCH REPORT 



Intc nat Application No 

PCl/bB 03/02804 



C.(Contlnuation) DOCUMENTS CONSIDERED TO BE RELEVANT 


Category ° 


Citation of document, with indication, where appropriate, of the relevant passages 


Relevant to claim No. 






111,112 


Y 


page 7349 


27,28, 






36-38, 






40, 






47-56, 






64-67, 






70-72, 






111,112 




page 7349 


X 


WO 00/29004 A (PEPT0R LTD ;PLAKSIN DANIEL 


22,23, 




(ID) 25 May 2000 (2000-05-25) 


25, 






29-31, 






38,40, 






47-56, 






61-63, 






70-72, 






111,112 




example 2 


X 


REITER Y ET AL: "An antibody 


22,23, 




single-domain phage display library of a 


25, 




native heavy chain variable region: 


29-31, 




isolation of functional single-domain VH 


38,40, 




molecules with a unique interface" 


47-56, 




JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, 


61—63 , 




vol. 290, no. 3, 


70-72, 




l January 1999 (1999-01-01), pages 


1*1*1 11 <*s 

111 ,112 




685-698, XP004461990 




ISSN: 0022-2836 






page 687 




P , A 


WO 03/002609 A (MEDICAL RES COUNCIL 


41,42, 




;T0MLINSON IAN (GB); WINTER GREG (GB); 


82 ? 83 




IGNATOV) 9 January 2003 (2003-01-09) 




page 29, line 8-15 






example 6 




P,X 


WO 03/035694 A (MUYLDERrlANS SERGE ; vLAAMS 


22, 113 




INTEKUNI V INST BIOTECH (BE)) 




"1 tn ~. * , onn'i / Anno r*i~ r* -i \ 

1 May 2003 (2003-05-01) 






page 21; example 6 




Y 


W0 97/30084 A (GENETICS INST) 


24-26, 




21 August 1997 (1997-08-21) 


32-35, 






38,40, 






47-56, 






64-67, 






70-72 , 






111,112 




example 6 




-/— 





Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



page 2 of 5 



INTERNATIONAL SEARCH REPORT 



tnt< anal Application No 

PL . , iB 03/02804 



C.(Continuatlon) DOCUMENTS CONSIDERED TO BE RELEVANT 


Category 0 


Citation of document, with indication, where appropriate, of the relevant passages 


Relevant to claim No. 


v 

X 


Lr U oOo Oo4 A ^MtUlLAL Kto LUUNL1L; 


9Q 

oy , 




i/r Mr*\/ i oon Moon nci 

io nay iyyu v,iyyu-ub— 10; 


A 7 E£ 

4/— bo , 




111 1 1 9 
1 1 1 , 1 1£ 


Y 


example y 


O A OC 






o^-ob , 






o o /in 
oo ,4U , 






A 7 C£ 

4/-bo , 






04-0/ , 






70-72, 






111,112 


Y 


example b 


LI ,Zo\ 






oo-oo, 






4U 3 






4/-50, 






o4-67, 






7r\ 7 0 






111 119 
111, 1 l£ 


Y 


— — _ 

WO 98/40469 A (MCFARLAND CLIVE DAVID 


27,28, 




. 1 IMnTDLIAnn datdtpta ammc /aii>. r&DHT at rpf» 
; UNUtKWUUD r A 1 Kit 1 A ANNh t AU J ; UAKU1AL LKL 


oO— oo , 




Nj 1/ oeptemoer iyyo \, iyyo-uy-i/ j 


/in 
4U, 






/17 C£ 

4/— bo , 






04—0/ , 






7fl— 79 
/ U"~/ c , 






111 119 
111,11c 




page o 




A 


SMITH BRYAN J ET AL: "Prolonged in vivo 


1-21, 




residence times or antioody fragments 


4/— bo , 




associated witn aiDumin 


£H 111 

OU ,111, 




D T Af AM 11 IP A TIT PUrMTCTDV 


119 

1 1*1 




vol. iz, no. o ? September /uui (.zuui-uyj, 






»~ ... rt.^. r» 7CA 7C£ VDnn997n7^ 1 

pacjes /bU-/bo 5 ArUu&£/u/ol 






ISdN: 1U4d-1oUZ 






tne wnoie document 




ft 

A 


VAN uhiv DLutKhrJ 1 ti AL: Duiiuing novel 


1— cl , 




binding Ugands to B7.1 and B7.2 based on 


47-56, 




human antibody single variable light chain 


60,111, 




domains" 


112 




JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, 






vol. 310, no. 3, 






13 July 2001 (2001-07-13), pages 591-601, 






XP004464206 






ISSN: 0022-2836 






abstract 






-/— 





Form PCT/ISA7210 (coniinuation of second sheet) (January 2004] 



oaae 3 of 5 



INTERNATIONAL SEARCH REPORT 



Inte >na! Application No 

PC./JB 03/02804 



C.(Continuation) DOCUMENTS CONSIDERED TO BE RELEVANT 


Category ° 


Citation of document, with indication, where appropriate, of the relevant passages 


Relevant to claim No. 


A 


■ a* ■ ■ \ # 1 f"*v ft ■ ft ft 1 B^H B^BV m » P * • B BY T • ~1 ■ * 

MUYLDERMANS S ET AL: UNIQUE 


1-21, 




SINGLE-DOMAIN ANTIGEN BINDING FRAGMENTS 

J* l ft ■%■» ftp' ^/ 1 If l •** ■ ^ * l • ft l *4U *— . 1 V 1^ aJU I % law* Jtm 1 ft wl 1 I ft 1 1 ftp! ■ 1 • ■ 1 ftp* 


47-56 

1 / aV V/ , 




DERIVES FROM NATURALLY OCCURRING CAMEL 


60,111, 




* ■ tP"* A 1 ||f i|J| VMK » | ft ft ■ w^"» PVP ft** PK VIM |M -a- a a 

HEAVY-CHAIN ANTIBODIES" 


112 




JOURNAL OF MOLECULAR RECOGNITION, HEYDEN & 






-h. A. ft | 1 Mi Vft. | _a**a, ft » pK W^ ft * jk PK 

SON LTD. , LONDON , GB , 






vol. 12, no. 2, March 1999 (1999-03), 






pages 131-140, XP009012180 






ISSN: 0952-3499 

>!■ S»* 1 » • ftp* -p* W 4— 1 *p» «pr 






abstract 




A 


RIECHMANN L ET AL: "Single domain 


1-21, 




antibodies: Comparison of camel VH and 


47-56, 




Pa p. a a * a * ■ ■ a- 

camel ised human VH domains" 


60,111, 




BIOSIS, 


112 




XP004187632 






the whole document 




A 


HOLLIGER P ET AL: "RETARGETING SERUM 


1-21, 




■as a ■ a a 1 afth ^Bk — a aa. a la ppa ■> * ■ vaft bbbb a ■ aaw am — aaM ^a. aaa* «■> hi — ass aa aa. ^bk Ml aai aaaa ^Ba. ■ ■ 

IMMUNOGLOBULIN WITH BISPECIFIC DIABODIES 


47-56, 




| 1 M ■» ■ | 1 *■* P— b*k Ban _«a_ T* f™ 1 1 ft 1 1 -~s A \ / ft ft At -ft** | a K. paaa T . — , a s— a a -w- ft K A 

NATURE BIOTECHNOLOGY, NATURE PUBLISHING, 


60,111, 




US, 


112 




vol. 15, July 1997 (1997-07), pages 






632-636, XP002921893 






*T» i**a a^%> El 1 <M ^a. — T <PV f« p— a 

ISSN: 1087-0156 






abstract 




A 


US 5 644 034 A (RATHJEN DEBORAH ANN ET 


22 23 




AL) 1 July 1997 (1997-07-01) 


25, 






29-31, 






38,40, 






47-56, 






61-63, 






70-72, 






111,112 




column 2 




A 


WO 91/02078 A (PEPTIDE TECHNOLOGY LTD) 

■ ■ ft^ ah g *■ ~* # ftBP" • 1 \ 1 tpM | 1 Bate lw Lai i J HP* ftp" ■ lift ^af ftaa XaF ^P« I Ea— 1 «— ' # 


22 23 




21 February 1991 (1991-02-21) 


25, 






29-31, 






38,40, 






47-56, 






61-63, 






70-72, 






111,112 




page 4, line 3,19 




-/-- 





Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



page 4 of 5 



INTERNATIONAL SEARCH REPORT 



In 'ionat Application No 

PCT/GB 03/02804 



C.(Continuation) DOCUMENTS CONSIDERED TO BE RELEVANT 



Category 



Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



A 



P,A 



TANHA J ET AL: "Optimal design features 
of camel i zed human single-domain antibody 
1 i for sir i g s rf 

THE JOURNAL OF BIOLOGICAL CHEMISTRY. 

UNITED STATES 6 JUL 2001, 

vol . 276, no. 27, 

6 July 2001 (2001-07-06), pages 

24774-24780, XP002283749 

ISSN: 0021-9258 

the whole document 

WO 02/072141 A (HERMAN WILLIAM) 
19 September 2002 (2002-09-19) 
the whole document 



1-117 



1-117 



Form PCT/tSA/210 (continuation of second sheet) (January 2004-) 



page 5 of 5 



INTERNATIONAL SEARCH REPORT 



srnational application No. 

PCT/GB 03/02804 



Box I Observations where certain claims were found unsearchable (Continuation of item 1 of first sheet) 



This international Search Report has not been established in respect of certain claims under Article 17(2) (a) for the following reasons: 
1. Claims Nos.: 

because they relate to subject matter not required to be searched by this Authority, namely: 



2. 



Claims Nos.: 

because they relate to parts of the International Application that do not comply with the prescribed requirements to such 
an extent that no meaningful International Search can be carried out, specifically: 



3. I | Claims Nos.: 

because they are dependent claims and are not drafted in accordance with the second and third sentences of Rule 6.4(a). 

Box (I Observations where unity of invention is Sacking (Continuation of item 2 of first sheet) 

This International Searching Authority found multiple inventions in this international application, as follows: 

see additional sheet 



X 



As ali required additional search fees were timely paid by the applicant, this International Search Report covers all 
searchable claims. 



2. | | As all searchable claims could be searched without effort justifying an additional fee, this Authority did not invite payment 
of any additional fee. 



3. | 1 As only some of the required additional search fees were timely paid by the applicant, this International Search Report 
' 1 covers only those claims for which fees were paid, specifically claims Nos.: 



4. | | No required additional search fees were timely paid by the applicant. Consequently, this International Search Report is 
restricted to the invention first mentioned in the claims; it is covered by claims Nos.: 



Remark on Protest | | The additional search fees were accompanied by the applicant's protest. 

[ X | No Protest accompanied the payment of additional search fees. 



Form PCT/ISA/210 (continuation of first sheet (1)) (July 1998) 



Internationa! Application No. PCT7 GB 03/02804 



FURTHER INFORMATION CONTINUED FROM PCT/ISA/ 210 



This International Searching Authority found multiple (groups of) 
inventions in this international application, as follows: 

1. Claims: 1-7,8-10,11-21,47-56 (all 10 in part), 

60 (in part), 111-112 (both in part) 



Ligand comprising a first immunoglobulin variable domain 
having a first antigen or epitope specificity and a second 
immunoglobulin variable domain having a second antigen or 
epitope specificity wherein one or both of the antigens or 
epitopes acts to increase the half-life of the ligand in 
vivo, and the variable domains are not complementary to one 
another. 



2. Claims: 22,23,25,29-31, 38 (in part) ,40, 

47-56 (all 10 part), 61-63, 70-72 (all 3 in part), 
111-112 (both in part) 



Single domain antibody monomer ligand specific for TNF- . 



3. Claims: 24,26,25,32-35, 38 (in part), 40, 

47-56 (all 10 in part), 64-67, 
70-72 (all 3 in part), 111-112 (both in part) 



Single domain antibody monomer ligand specific for TNF 
receptor 1. 



4. Claims: 27, 28, 36,37, 38 (in part)40, 

47-56 (all 10 in part), 64-67, 
70-72 (all 3 in part), 111-112 (both in part) 



Single domain antibody monomer ligand specific for serum 
albumin. 



5. Claims: 39, 47-56 (all 10 in part), 

111-112 (both in part). 



Single domain antibody monomer ligand comprising a terminal 
Cys residue. 



6. Claims: 41-46, 47-56 (all 10 in part), 60 (in part). 

A dual specific ligand comprising at least a single domain 
antibody monomer according to inventions 2-5. 



page 1 of 2 



International Application No. PC7Y GB 03/02804 



FURTHER INFORMATION CONTINUED FROM PCT/ISA/ 210 



7. Claims: 73-81, 82-104, 105-110 

A closed conformation multi specif ic ligand comprising a 
first epitope binding domain having a first epitope binding 
specificity and a non-complementary second epitope binding 
domain having a second epitope binding specificity, wherein 
the first and second binding specificities are capable of 
competing for epitope binding such that the closed 
conformation multi-specific ligand cannot bind both 
epitopes simultaneously. Methods for producing the closed 
conformation multi specific ligand. Methods using the closed 
conformation multi specif ic ligands. 



8. Claims: 113-117 

Method for preparing a chelating mul timeric ligand, specific 
for adjacent epitopes. 



page 2 of 2 



INTERNATIONAL SEARCH REPORT 

Information on patent family members 



lr itional Application No 

PCl/GB 03/02804 



Patent document 


Publication 




Patent family 




Publication 


cited in search report 


date 




member(s) 




date 


WO 0029004 A 


25-05-2000 


AU 


765201 


B2 


11-09-2003 




AU 


6486999 


A 


05-06-2000 




a a 

CA 


2351669 


A 1 

Al 


P> p ftr p>/"> p> r> 

25-05-2000 




EP 


1131079 


Al 


12-09-2001 




WO 


0029004 


Al 


25-05-2000 




KIT 

HZ 


IT "1 *f A fC /2 

511466 


A 


O P\ P> A o o r\ o 

29-04-2003 




US 


2002012909 


Al 


31-01-2002 


W0 03002609 A 


09-01-2003 


r\ a 

CA 


2447851 


A 1 

Al 


09-01-2003 




EP 


1399484 


ft A 

A2 


24-03-2004 




WO 


AOAftft/^Aft 

03002609 


ft a 

A2 


r\r\ a *i oaao 

09-01-2003 


W0 03035694 A 


01-05-2003 


EP 


1306386 


n *i 
Al 


02-05-2003 




WO 


ft ft i - p* P"l >| 

03035694 


A2 


ft"i ftr" •ftftrtft 

01-05-2003 


W0 9730084 A 


21-08-1997 


us 


5843675 


A 


01-12-1998 




1 1 a 

US 


r ~j h ft ft O 1 

5712381 


A 


27-01-1998 




All 

AU 


O ft ft /"V *T 

2268697 


A 


ft ft Art *1 A ft T 

02-09-1997 




WO 


9730084 


Al 


fti p\ p\ -i pv pv —y 

21-08-1997 




1 IP 

us 


6322972 


Bl 


A"7 11 AAAt 

27-11-2001 




us 


P" P"v pv -1 p"? r 

5891675 


ii 
A 


06-04-1999 




1 IP 

us 


2002164716 


Al 


AT f "1 AAAA 

07-11-2002 


EP 0368684 A 


16-05-1990 


ft T* 

AT 


"1 ft P> P** O "1 

102631 


T 


1 r* AO "f AA A 

15-03-1994 




a l l 

AU 


634186 


D O 
D£ 


1 A AO 1AAA 

18-02-1993 




nil 

AU 


4520189 


A 


AA A r~ 1 AAA 

28-05-1990 




CA 


ps p\ ^ p« p» p* pi 
2002868 


a -i 
Al 


"1 "1 A r~ "1 AAA 

11-05-1990 




DE 


>C f> P^i "I o p* r* P> 

689 I 3658 


Dl 


1/1 ft Jt 1 ftft A 

14-04-1994 




rv r" 

DE 


pop ^ « p» r" ft 

68913658 


T p> 

T2 


p» p» p\ py "lAftil 

08-09-1994 




DK 


164790 


A 


ft "T ft Pv "J Aft/"» 

07-09-1990 




EP 


ft o /- t~\ f A n 

0368684 


Al 


1 fT A P* "1 AAA 

16-05-1990 




ES 


nnp rtPT7 

2052027 


T O 

T3 


ft -1 ft"T 1Aft^ 

01-07-1994 




wo 


9005144 


Al 


*IT AP* 1 AAA 

17-05-1990 




JP 


2919890 


B2 


19-07-1999 




JP 


3502801 


T 


27-06-1991 




KR 


184860 


D 1 


A "1 A /> 1 AAA 

01-04-1999 




MO 


903059 


A 


07-09-1990 




US 


2003130496 


Al 


10-07-2003 




US 


pi pv r> o *i "i yi /*rA 

2003114659 


ft -1 

Al 


"1 A ft /T OAAA 

19-06-2003 




us 


p* pi p> r~ i p* 

6248516 


Bl 


19-06-2001 




us 


/p r— « t— -i /J pj 

6545142 


Bl 


r> p\ ft ft OAAA 

08-04-2003 


W0 9840469 A 


17-09-1998 


All 

AU 


P P P P "T A A 

6602798 


A 


29-09-1998 




1 1 o 
WO 


9840469 


Al 


-i — r pi p> i p\ p\ pv 

17-09-1998 


US 5644034 A 


01-07-1997 


us 


pi pn <*\ w oft.r""n 

2003139577 


Al 


ftft r\'~~l A A A A 

24-07-2003 




1 1 a 

us 


ft p» p> ft -1 r\ AT O ft 

2003139580 


Al 


AVI AT AAAO 

24-07-2003 




us 


ftftftA 1 

2003162948 


Al 


AA AA OAAO 

28-08-2003 




1 1 o 

us 


O AAO 1 AT AOA 

2003135029 


A "I 

Al 


1 *T A*7 OAAO 

17-07-2003 




1 J o 

us 


A AAA 1 PP p TP 

2003199678 


Al 


AA 1 A OAAO 

23-10-2003 




us 


2003232970 


Al 


18-12-2003 




us 


2003170204 


Al 


11-09-2003 




us 


2003208049 


Al 


06-11-2003 




us 


2004002588 


Al 


01-01-2004 




us 


2003171553 


Al 


11-09-2003 




us 


2003166874 


Al 


04-09-2003 




us 


2003216552 


Al 


20-11-2003 




us 


2004002589 


Al 


01-01-2004 



Form PCT/ISA/210 (patent family annex) (January 2004) 



page 1 of 3 



INTERNATIONAL SEARCH REPORT 

Information on patent family members 



| onal Application No 

, 3B 03/02804 



Patent document 




Publication 




Patent family 




Publication 






date 




memberfs^ 




date 


US 5644034 


A 


US 


2003208047 


Al 


06-11-2003 






US 


2003171554 


Al 


11-09-2003 






US 


2003171555 


Al 


11-09-2003 






US 


2004002590 


Al 


01-01-2004 






US 


2004092721 


Al 


13-05-2004 






US 


2003225254 


Al 


04-12-2003 






US 


6416757 


Bl 


09-07-2002 






US 


2003232971 


Al 


18-12-2003 






US 


6593458 


Bl 


15-07-2003 






US 


2001018507 


Al 


30-08-2001 






US 


2001023287 


Al 


20-09-2001 






US 


2001018508 


Al 


30-08-2001 






US 


5959087 


A 


28-09-1999 






AU 


640400 


B2 


26-08-1993 






AU 


6145490 


A 


11-03-1991 






WO 


9102078 


Al 


21-02-1991 






CA 


2064915 


Al 


08-02-1991 






DE 


690Z7121 


Dl 


ci— uo-iyyo 






DE 


69027121 


T2 


14-11-1996 






DK 


486526 


T3 


24-06-1996 






EP 


0486526 


Al 


27-05-1992 






JP 


3443119 


B2 


02-09-2003 






JP 
JP 


2003096096 
2003231699 


A 

A 


03-04-2003 
19-08-2003 


WO 9102078 


A 


21-02-1991 


AU 


640400 


B2 


26-08-1993 






AU 


6145490 


A 


11-03-1991 






WO 


9102078 


Al 


21-02-1991 






EP 


0486526 


Al 


27-05-1992 






CA 


2064915 


Al 


08-02-1991 






DE 


69027121 


Dl 


27-06-1996 






DE 


69027121 


T2 


14-11-1996 






DK 


486526 


T3 


24-06-1996 






JP 


3443119 


B2 


02-09-2003 






JP 


2003096096 


A 


03-04-2003 






JP 


2003231699 


A 


19-08-2003 






US 


2003162948 


Al 


28-08-2003 






US 


2003232970 


Al 


18-12-2003 






US 


2004002588 


Al 


01-01-2004 






US 


2003171553 


Al 


11-09-2003 






US 


2003166874 


Al 


04-09-2003 






US 


2003216552 


Al 


20-11-2003 






US 


2004002589 


Al 


01-01-2004 






US 


2003208047 


Al 


06-11-2003 






US 


5644034 


A 


01-07-1997 






US 


2003171554 


Al 


11-09-2003 






US 


2003171555 


Al 


11-09-2003 






US 


2004002590 


Al 


01-01-2004 






us 


2004092721 


Al 


13-05-2004 












U4— id.—cUU o 






us 


6416757 


Bl 


09-07-2002 






us 


2003232971 


Al 


18-12-2003 






us 


6593458 


Bl 


15-07-2003 






us 


2001018507 


Al 


30-08-2001 






us 


2001023287 


Al 


20-09-2001 






us 


2001018508 


Al 


30-08-2001 


W0 02072141 


A 


19-09-2002 


wo 


02072141 


A2 


19-09-2002 



Form PCT/ISA/210 (patent family annex) (January 2004) 



page 2 of 3 



INTERNATIONAL SEARCH REPORT 

iformation on patent family members 



fn ional Application No 

Pui/fiB 03/02804 



Patent document 
cited in search report 



Publication 
date 



Patent family 
member(s) 



Publication 
date 



WO 02072141 



A 



WO 



03057732 A2 



17-07-2003 



Form PCT/ISA/210 (patent family annex) (January 2004) 



page 3 of 3 



(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



CORRECTED VERSION 



(19) World Intellectual Property Organization 

International Bureau 

(43) International Publication Date 
8 January 2004 (08.01.2004) 





PCT 



(10) International Publication Number 

WO 2004/003019 A3 



(51) International Patent Classification : C07K 16/24, 

16/18, 16/28, A61K 39/395, C07K 16/46, C12N 15/63, 
15/62, 15/13 

(21) International Application Number: 

PCT/GB2003/002804 

(22) International Filing Date: 30 June 2003 (30.06.2003) 



(25) Filing Language: 

(26) Publication Language: 



English 
English 



(30) Priority Data: 

PCT/GB02/03014 28 June 2002 (28.06.2002) GB 
0230202.4 27 December 2002 (27.12.2002) GB 

(7 1) Applicant (fo r all designated States except US ) : DOMAN- 
TIS LIMITED [GB/GB]; 315 Cambridge Science Park, 
Cambridge, Cambridgeshire CB4 0WG (GB). 

(72) Inventors; and 

(75) Inventors/Applicants (for US only): WINTER, Greg 
[GB/GB]; MRC Laboratory of Molecular Biology, Hills 
Road, Cambridge CB2 2QH (GB). TOMLINS ON, Ian 
[GB/GB]; Domantis Limited, Granta Park, Abington, 
Cambridge CB1 6GS (GB). IGNATOVICH, Olga 
[BY/GB]; Domantis Limited, Granta Park, Abington, 
Cambridge CB1 6GS (GB). HOLT, Lucy [GB/GB]; 
Domantis Limited, Granta Park, Abington, Cambridge 
CB1 6GS (GB). DE ANGELIS, Elena [IT/GB]; Domantis 
Limited, Granta Park, Abington, Cambridge CB1 6GS 
(GB). JONES, Phillip [GB/GB]; 15 Impington Lane, 
Impington, Cambridge CB4 9LT (GB). 

(74) Agents: MASCHIO, Antonio et al.; D. Young & Co., 120 
Holborn, London EC1N 2DY (GB). 



(81) Designated States (national): AE, AG, AL, AM, AT, AU, 
AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CO, CR, CU, 
CZ, DE, DK, DM, DZ, EC, EE, ES, FT, GB, GD, GE, GH, 
GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, 
LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, 
MX, MZ, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, 
SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, 
UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. 

(84) Designated States (regional): ARIPO patent (GH, GM, 
KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZM, ZW), 
Eurasian patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, 
TM), European patent (AT, BE, BG, CH, CY, CZ, DE, 
DK, EE, ES, FI, FR, GB, GR, HU, IE, IT, LU, MC, NL, 
PT, RO, SE, SI, SK, TR), OAPI patent (BF, BJ, CF, CG, 
CI, CM, GA, GN, GQ, GW, ML, MR, NE, SN, TD, TG). 

Declaration under Rule 4.17: 

— of invento rship ( Rule 4. 1 7( iv ) ) 

Published: 

— with international search report 

(88) Date of publication of the international search report: 

10 September 2004 

(48) Date of publication of this corrected version: 

27 April 2006 

(15) Information about Correction: 

see PCT Gazette No. 17/2006 of 27 April 2006 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



< 

OS 



(54) Title: IMMUNOGLOBIN SINGLE VARIANT ANTIGEN-BINDING DOMAINS AND DUAL-SPECIFIC CONSTRUCTS 

(57) Abstract: The invention provides a dual-specific ligand comprising a first immunoglobulin variable domain having a first 
binding specificity and a complementary or non-complementary immunoglobulin variable domain having a second binding speci- 
ficity. 



o 



WO 2004/003019 



PCT/GB2003/002804 



Ligand 

The present invention relates to dual specific ligands. In particular, the invention 
provides a method for the preparation of dual-specific ligands comprising a first 
5 immunoglobulin single variable domain binding to a first antigen or epitope, and a second 
immunoglobulin single variable domain binding to a second antigen or epitope. More 
particularly, the invention relates to dual-specific ligands wherein binding to at least one 
of the first and second antigens or epitopes acts to increase the half-life of the ligand in 
vivo. Open and closed conformation ligands comprising more than one binding specificity 
10 are described. 

Introduction 

The antigen binding domain of an antibody comprises two separate regions: a heavy 

15 chain variable domain (Vr) an d a. light chain variable domain (v L : which can be either 
V K or Vx,). The antigen binding site itself is formed by six polypeptide loops: three from 
V H domain (HI, H2 and H3) and three from v L domain (LI, L2 and L3). A diverse 
primary repertoire of V genes that encode the Vh an d Vl domains is produced by the 
combinatorial rearrangement of gene segments. The Vh g ei * e is produced by the 

20 recombination of three gene segments, Vh> ^ anc * J H- hi humans, there are approximately 
51 functional Vh segments (Cook and Tomlinson (1995) Immunol Today, 16: 237), 25 
functional D segments (Corbett et ah (1997) J. Moh Bioh, 268: 69) and 6 functional Jh 
segments (Ravetch et ah (1981) Cell, 27: 583), depending on the haplotype. The Vh 
segment encodes the region of the polypeptide chain which forms the first and second 

25 antigen binding loops of the Vh domain (HI and H2), whilst the y H , D an d ^H segments 
combine to form the third antigen binding loop of the Vh domain (H3). The Vl g e ^e is 
produced by the recombination of only two gene segments, Vl an d JL- hi humans, there 
are approximately 40 functional V K segments (Schable and Zachau (1993) Bioh Chem. 
Hoppe-Seyler, 374: 1001), 31 functional Y% segments (Williams et ah (1996) J. Moh 

30 Bioh, 264: 220; Kawasaki et ah (1997) Genome Res., 7: 250), 5 functional J K segments 
(Hieter et ah (1982) J. Bioh Chem., 257: 1516) and 4 functional segments (Vasicek 



WO 2004/003019 PCT/GB2003/002804 

2 

and Leder (1990) J. Exp. Med., 172: 609), depending on the haplotype. The v L segment 
encodes the region of the polypeptide chain which forms the first and second antigen 
binding loops of the v L domain (LI and L2), whilst the v L and J L segments combine to 
form the third antigen binding loop of the v L domain (L3). Antibodies selected from this 
5 primary repertoire are believed to be sufficiently diverse to bind almost all antigens with 
at least moderate affinity. High affinity antibodies are produced by "affinity maturation" 
of the rearranged genes, in which point mutations are generated and selected by the 
immune system on the basis of improved binding. 

10 Analysis of the structures and sequences of antibodies has shown that five of the six 
antigen binding loops (HI, H2, LI, L2, L3) possess a limited number of main-chain 
conformations or canonical structures (Chothia and Lesk (1987) J, Mol Biol, 196: 901; 
Chothia et al (1989) Nature, 342: 877). The main-chain conformations are determined by 
(i) the length of the antigen binding loop, and (ii) particular residues, or types of residue, 

15 at certain key position in the antigen binding loop and the antibody framework. Analysis 
of the loop lengths and key residues has enabled us to the predict the main-chain 
conformations of HI, H2, LI, L2 and L3 encoded by the majority of human antibody 
sequences (Chothia et al (1992) J. Mol Biol, 221: 799; Tomlinson et al (1995) EMBO 
J., 14: 4628; Williams et al (1996) J. Mol Biol, 264: 220). Although the H3 region is 

20 much more diverse in terms of sequence, length and structure (due to the use of D 
segments), it also forms a limited number of main-chain conformations for short loop 
lengths which depend on the length and the presence of particular residues, or types of 
residue, at key positions in the loop and the antibody framework (Martin et al (1996) J. 
Mol Biol, 263: 800; Shirai et al (1996) FEBS Letters, 399: 1. 

25 

Bispecific antibodies comprising complementary pairs of v H and V L regions are known in 
the art. These bispecific antibodies must comprise two pairs of v H and V L S > eaclx Vi/Vl 
pair binding to a single antigen or epitope. Methods described involve hybrid hybridomas 
(Milstein & Cuello AC, Nature 305:537-40), minibodies (Hu et al, (1996) Cancer Res 
30 56:3055-3061;), diabodies (Holliger et al, (1993) Proc. Natl. Acad. Sci. USA 90, 6444- 
6448; WO 94/13804), chelating recombinant antibodies (CRAbs; (Neri et al, (1995) J. 
Mol. Biol. 246, 367-373), biscFv (e.g. Atwell et al, (1996) Mol. Immunol. 33, 1301- 
1312), "knobs in holes" stabilised antibodies (Carter et al, (1997) Protein Sci. 6, 781- 



WO 2004/003019 PCT/GB2003/002804 

3 

788). In each case each antibody species comprises two antigen-binding sites, each 
fashioned by a complementary pair of v H an d V L domains. Each antibody is thereby able 
to bind to two different antigens or epitopes at the same time, with the binding to EACH 
antigen or epitope mediated by a Vh an d its complementary Vl domain. Each of these 
5 techniques presents its particular disadvantages; for instance in the case of hybrid 
hybridomas, inactive V u /y L P airs can greatly reduce the fraction of bispecific IgG. 
Furthermore, most bispecific approaches rely on the association of the different wVl 
pairs or the association of Vh an d V L chains to recreate the two different Vh^Vl binding 
sites. It is therefore impossible to control the ratio of binding sites to each antigen or 
10 epitope in the assembled molecule and thus many of the assembled molecules will bind 
to one antigen or epitope but not the other. In some cases it has been possible to engineer 
the heavy or light chains at the sub-unit interfaces (Carter et al, 1997) in order to improve 
the number of molecules which have binding sites to both antigens or epitopes but this 
never results in all molecules having binding to both antigens or epitopes. 

15 

There is some evidence that two different antibody binding specificities might be 
incorporated into the same binding site, but these generally represent two or more 
specificities that correspond to structurally related antigens or epitopes or to antibodies 
that are broadly cross-reactive.. For example, cross-reactive antibodies have been 

20 described, usually where the two antigens are related in sequence and structure, such as 
hen egg white lysozyme and turkey lysozyme (McCafferty et al., WO 92/01047) or to 
free hapten and to hapten conjugated to carrier (Griffiths AD et al. EMBO J 1994 13:14 
3245-60). In a further example, WO 02/02773 (Abbott Laboratories) describes antibody 
molecules with "dual specificity". The antibody molecules referred to are antibodies 

25 raised or selected against multiple antigens, such that their specificity spans more than a 
single antigen. Each complementary Vh/V l P air in the antibodies of WO 02/02773 
specifies a single binding specificity for two or more structurally related antigens; the v H 
and Vl domains in such complementary pairs do not each possess a separate specificity. 
The antibodies thus have a broad single specificity which encompasses two antigens, 

30 which are structurally related. Furthermore natural autoantibodies have been described 
that are polyreactive (Casali & Notkins, Ann. Rev. Immunol. 7, 515-531), reacting with at 
least two (usually more) different antigens or epitopes that are not structurally related. It 



WO 2004/003019 PCT/GB2003/002804 

4 

has also been shown that selections of random peptide repertoires using phage display 
technology on a monoclonal antibody will identify a range of peptide sequences that fit 
the antigen binding site. Some of the sequences are highly related, fitting a consensus 
sequence, whereas others are very different and have been termed mimotopes (Lane & 
Stephen, Current Opinion in Immunology, 1993, 5, 268-271). It is therefore clear that a 
natural four-chain antibody, comprising associated and complementary Vh and y L 
domains, has the potential to bind to many different antigens from a large universe of 
known antigens. It is less clear how to create a binding site to two given antigens in the 
same antibody, particularly those which are not necessarily structurally related. 

Protein engineering methods have been suggested that may have a bearing on this. For 
example it has also been proposed that a catalytic antibody could be created with a 
binding activity to a metal ion through one variable domain, and to a hapten (substrate) 
through contacts with the metal ion and a complementary variable domain (Barbas et al., 
1993 Proc. Natl. Acad. Sci USA 90, 6385-6389). However in this case, the binding and 
catalysis of the substrate (first antigen) is proposed to require the binding of the metal ion 
(second antigen). Thus the binding to the Vf/V L pairing relates to a single but multi- 
component antigen. 

Methods have been described for the creation of bispecific antibodies from camel 
antibody heavy chain single domains in which binding contacts for one antigen are 
created in one variable domain, and for a second antigen in a second variable domain. 
However the variable domains were not complementary. Thus a first heavy chain variable 
domain is selected against a first antigen, and a second heavy chain variable domain 
against a second antigen, and then both domains are linked together on the same chain to 
give a bispecific antibody fragment (Conrath et al., J. Biol. Chem. 270, 27589-27594). 
However the camel heavy chain single domains are unusual in that they are derived from 
natural camel antibodies which have no light chains, and indeed the heavy chain single 
domains are unable to associate with camel light chains to form complementary y H and 
V L pairs. 

Single heavy chain variable domains have also been described, derived from natural 
antibodies which are normally associated with light chains (from monoclonal antibodies 



WO 2004/003019 PCT/GB2003/002804 

5 

or from repertoires of domains; see EP-A-0368684). These heavy chain variable 
domains have been shown to interact specifically with one or more related antigens but 
have not been combined with other heavy or light chain variable domains to create a 
ligand with a specificity for two or more different antigens . Furthermore, these single 
5 domains have been shown to have a very short in vivo half-life. Therefore such domains 
are of limited therapeutic value. 

It has been suggested to make bispecific antibody fragments by linking heavy chain 
variable domains of different specificity together (as described above). The disadvantage 

10 with this approach is that isolated antibody variable domains may have a hydrophobic 
interface that normally makes interactions with the light chain and is exposed to solvent 
and may be "sticky" allowing the single domain to bind to hydrophobic surfaces. 
Furthermore, in the absence of a partner light chain the combination of two or more 
different heavy chain variable domains and their association, possibly via their 

1 5 hydrophobic interfaces, may prevent them from binding to one in not both of the ligands 
they are able to bind in isolation. Moreover, in this case the heavy chain variable 
domains would not be associated with complementary light chain variable domains and 
thus may be less stable and readily unfold (Worn & Pluckthun, 1998 Biochemistry 37, 
13120-7). 

20 

Summary of the invention 

The inventors have described, in their copending international patent application WO 
03/002609 as well as copending unpublished UK patent application 0230203.2, dual 
25 specific immunoglobulin ligands which comprise immunoglobulin single variable 
domains which each have different specificities. The domains may act in competition 
with each other or independently to bind antigens or epitopes on target molecules. 

In a first configuration, the present invention provides a further improvement in dual 
30 specific ligands as developed by the present inventors, in which one specificity of the 
ligand is directed towards a protein or polypeptide present in vivo in an organism which 
can act to increase the half-life of the ligand by binding to it. 



WO 2004/003019 PCT/GB2003/002804 

6 

Accordingly, in a first aspect, there is provided a dual-specific ligand comprising a first 
immunoglobulin single variable domain having a binding specificity to a first antigen or 
epitope and a second complementary immunoglobulin single variable domain having a 
binding activity to a second antigen or epitope, wherein one or both of said antigens or 
epitopes acts to increase the half-life of the ligand in vivo and wherein said first and 
second domains lack mutually complementary domains which share the same specificity, 
provided that said dual specific ligand does not consist of an anti-HS A V H domain and an 
anti-p galactosidase V K domain. Preferably, that neither of the first or second variable 
domains binds to human serum albumin (HSA). 

Antigens or epitopes which increase the half-life of a ligand as described herein are 
advantageously present on proteins or polypeptides found in an organism in vivo. 
Examples include extracellular matrix proteins, blood proteins, and proteins present in 
various tissues in the organism. The proteins act to reduce the rate of ligand clearance 
from the blood, for example by acting as bulking agents, or by anchoring the ligand to a 
desired site of action. Examples of antigens/epitopes which increase half-life in vivo are 
given in Annex 1 below. 

Increased half-life is useful in in vivo applications of immunoglobulins, especially 
antibodies and most especially antibody fragments of small size. Such fragments (Fvs, 
disulphide bonded Fvs, Fabs, scFvs, dAbs) suffer from rapid clearance from the body; 
thus, whilst they are able to reach most parts of the body rapidly, and are quick to produce 
and easier to handle, their in vivo applications have been limited by their only brief 
persistence in vivo. The invention solves this problem by providing increased half-life of 
the ligands in vivo and consequently longer persistence times in the body of the functional 
activity of the ligand. 

Methods for pharmacokinetic analysis and determination of ligand half-life will be 
familiar to those skilled in the art. Details may be found in Kenneth A et al: Chemical 
Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al 
Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to 
"Pharmacokinetics", M Gibaldi & D Perron, published by Marcel Dekker, 2 nd Rev. ex 



WO 2004/003019 PCT/GB2003/002804 

7 

edition (1982), which describes pharmacokinetic parameters such as t alpha and t beta 
half lives and area under the curve (AUC). 

Half lives (tVz alpha and Wi beta) and AUC can be determined from a curve of serum 
5 concentration of ligand against time. The WinNonlin analysis package (available from 
Pharsight Corp., Mountain View, CA94040, USA) can be used, for example, to model the 
curve. In a first phase (the alpha phase) the ligand is undergoing mainly distribution in 
the patient, with some elimination. A second phase (beta phase) is the terminal phase 
when the ligand has been distributed and the serum concentration is decreasing as the 

10 ligand is cleared from the patient. The t alpha half life is the half life of the first phase 
and the t beta half life is the half life of the second phase. Thus, advantageously, the 
present invention provides a ligand or a composition comprising a ligand according to the 
invention having a ta half— life in the range of 15 minutes or more. In one embodiment, 
the lower end of the range is 30 minutes, 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 

15 hours, 6 hours, 7 hours, 10 hours, 11 hours or 12 hours. In addition, or alternatively, a 
ligand or composition according to the invention will have a ta half life in the range of up 
to and including 12 hours. In one embodiment, the upper end of the range is 11, 10, 9, 8, 
7, 6 or 5 hours. An example of a suitable range is 1 to 6 hours, 2 to 5 hours or 3 to 4 
hours. 

20 

Advantageously, the present invention provides a ligand or a composition comprising a 
ligand according to the invention having a tp half— life in the range of 2.5 hours or more. 
In one embodiment, the lower end of the range is 3 hours, 4 hours, 5 hours, 6 hours, 7 
hours, 10 hours , 11 hours, or 12 hours. In addition, or alternatively, a ligand or 

25 composition according to the invention has a t(3 half-life in the range of up to and 
including 21 days. In one embodiment, the upper end of the range is 12 hours, 24 hours, 
2 days, 3 days, 5 days, 10 days, 15 days or 20 days. Advantageously a ligand or 
composition according to the invention will have a tp half life in the range 12 to 60 hours. 
In a further embodiment, it will be in the range 12 to 48 hours. In a further embodiment 

30 still, it will be in the range 12 to 26 hours. 

In addition, or alternatively to the above criteria, the present invention provides a ligand 
or a composition comprising a ligand according to the invention having an AUC 



WO 2004/003019 PCT/GB2003/002804 

8 

value (area under the curve) in the range of 1 mg.min/ml or more. In one embodiment, 
the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300mg.min/ml. In addition, or 
alternatively, a ligand or composition according to the invention has an AUC in the range 
of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 
5 200, 150, 100, 75 or 50 mg.min/ml. Advantageously a ligand according to the invention 
will have a AUC in the range selected from the group consisting of the following: 15 to 
150mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50mg.min/ml. 

In a first embodiment, the dual specific ligand comprises two complementary variable 
10 domains, i.e. two variable domains that, in their natural environment, are capable of 
operating together as a cognate pair or group even if in the context of the present 
invention they bind separately to their cognate epitopes. For example, the complementary 
variable domains may be immunoglobulin heavy chain and light chain variable domains 
(V H and V L ). V H and V L domains are advantageously provided by scFv or Fab antibody 
15 fragments. Variable domains may be linked together to form multivalent ligands by, for 
example: provision of a hinge region at the C-terminus of each V domain and disulphide 
bonding between cysteines in the hinge regions; or provision of dAbs each with a cysteine 
at the C-terminus of the domain, the cysteines being disulphide bonded together; or 
production of V-CH & V-CL to produce a Fab format; or use of peptide linkers (for 
20 example Gly 4 Ser linkers discussed hereinbelow) to produce dimers, trimers and further 
multimers. 

The inventors have found that the use of complementary variable domains allows the two 
domain surfaces to pack together and be sequestered from the solvent. Furthermore the 

25 complementary domains are able to stabilise each other. In addition, it allows the creation 
of dual-specific IgG antibodies without the disadvantages of hybrid hybridomas as used 
in the prior art, or the need to engineer heavy or light chains at the sub-unit interfaces. 
The dual-specific ligands of the first aspect of the present invention have at least one 
Vh/Vl pair. A bispecific IgG according to this invention will therefore comprise two 

30 such pairs, one pair on each arm of the Y-shaped molecule. Unlike conventional 
bispecific antibodies or diabodies, therefore, where the ratio of chains used is 
determinative in the success of the preparation thereof and leads to practical difficulties, 
the dual specific ligands of the invention are free from issues of chain balance. Chain 



WO 2004/003019 PCT/GB2003/002804 

9 

imbalance in conventional bi-specific antibodies results from the association of two 
different V L chains with two different V H chains, where V L chain 1 together with V H 
chain 1 is able to bind to antigen or epitope 1 and V L chain 2 together with V H chain 2 is 
able to bind to antigen or epitope 2 and the two correct pairings are in some way linked to 
5 one another. Thus, only when V L chain 1 is paired with V H chain 1 and V L chain 2 is 
paired with V H chain 2 in a single molecule is bi-specificity created. Such bi-specific 
molecules can be created in two different ways. Firstly, they can be created by association 
of two existing V H /V L pairings that each bind to a different antigen or epitope (for 
example, in a bi-specific IgG). In this case the V H /V L pairings must come all together in a 

10 1:1 ratio in order to create a population of molecules all of which are bi-specific. This 
never occurs (even when complementary CH domain is enhanced by "knobs into holes" 
engineering) leading to a mixture of bi-specific molecules and molecules that are only 
able to bind to one antigen or epitope but not the other. The second way of creating a bi- 
specific antibody is by the simultaneous association of two different Vh chain with two 

15 different V L chains (for example in a bi-specific diabody). In this case, although there 
tends to be a preference for V L chain 1 to pair with V H chain 1 and Vl chain 2 to pair with 
Vh chain 2 (which can be enhanced by "knobs into holes" engineering of the V L and V H 
domains), this paring is never achieved in all molecules, leading to a mixed formulation 
whereby incorrect pairings occur that are unable to bind to either antigen or epitope. 

20 

Bi-specific antibodies constructed according to the dual-specific ligand approach 
according to the first aspect of the present invention overcome all of these problems 
because the binding to antigen or epitope 1 resides within the Vh or Vl domain and the 
binding to antigen or epitope 2 resides with the complementary Vl or Vh domain, 
25 respectively. Since V H and V L domains pair on a 1:1 basis all V H /V L pairings will be bi- 
specific and thus all formats constructed using these V H /V L pairings (Fv, scFvs, Fabs, 
minibodies, IgGs etc) will have 100% bi-specific activity. 

In the context of the present invention, first and second "epitopes" are understood to be 
30 epitopes which are not the same and are not bound by a single monospecific ligand. In 
the first configuration of the invention, they are advantageously on different antigens, one 
of which acts to increase the half-life of the ligand in vivo. Likewise, the first and second 
antigens are advantageously not the same. 



WO 2004/003019 



PCT/GB2003/002804 



10 

The dual specific ligands of the invention do not include ligands as described in WO 
02/02773. Thus, the ligands of the present invention do not comprise complementary 
V H /V L pairs which bind any one or more antigens or epitopes co-operatively. Instead, the 
ligands according to the first aspect of the invention comprise a V h /Vl complementary 
pair, wherein the V domains have different specificities. 

Moreover, the ligands according to the first aspect of the invention comprise Vh/Vl 
complementary pairs having different specificities for non-structurally related epitopes or 
antigens. Structurally related epitopes or antigens are epitopes or antigens which possess 
sufficient structural similarity to be bound by a conventional V h /Vl complementary pair 
which acts in a co-operative manner to bind an antigen or epitope; in the case of 
structurally related epitopes, the epitopes are sufficiently similar in structure that they 
"fit" into the same binding pocket formed at the antigen binding site of the V H /V L dimer. 

In a second aspect, the present invention provides a ligand comprising a first 
immunoglobulin variable domain having a first antigen or epitope binding specificity and 
a second immunoglobulin variable domain having a second antigen or epitope binding 
specificity wherein one or both of said first and second variable domains bind to an 
antigen which increases the half-life of the ligand in vivo, and the variable domains are 
not complementary to one another. 

In one embodiment, binding to one variable domain modulates the binding of the ligand 
to the second variable domain. 

In this embodiment, the variable domains may be, for example, pairs of V H domains or 
pairs of Vl domains. Binding of antigen at the first site may modulate, such as enhance or 
inhibit, binding of an antigen at the second site. For example, binding at the first site at 
least partially inhibits binding of an antigen at a second site. In such an embodiment, the 
ligand may for example be maintained in the body of a subject organism in vivo through 
binding to a protein which increases the half-life of the ligand until such a time as it 
becomes bound to the second target antigen and dissociates from the half-life increasing 
protein. 



WO 2004/003019 



PCT/GB2003/002804 



11 

Modulation of binding in the above context is achieved as a consequence of the structural 
proximity of the antigen binding sites relative to one another. Such structural proximity 
can be achieved by the nature of the structural components linking the two or more 
5 antigen binding sites, eg by the provision of a ligand with a relatively rigid structure that 
holds the antigen binding sites in close proximity. Advantageously, the two or more 
antigen binding sites are in physically close proximity to one another such that one site 
modulates the binding of antigen at another site by a process which involves steric 
hindrance and/or conformational changes within the immunoglobulin molecule. 

10 

The first and the second antigen binding domains may be associated either covalently or 
non-covalently. In the case that the domains are covalently associated, then the 
association may be mediated for example by disulphide bonds or by a polypeptide linker 
such as (Gly 4 Ser) n , where n = from 1 to 8, eg, 2, 3, 4, 5 or 7. 

15 

Ligands according to the invention may be combined into non-immunoglobulin multi- 
ligand structures to form multivalent complexes, which bind target molecules with the 
same antigen, thereby providing superior avidity, while at least one variable domain binds 
an antigen to increase the half life of the multimer. For example natural bacterial 

20 receptors such as SpA have been used as scaffolds for the grafting of CDRs to generate 
ligands which bind specifically to one or more epitopes. Details of this procedure are 
described in US 5,831,012. Other suitable scaffolds include those based on fibronectin 
and affibodies. Details of suitable procedures are described in WO 98/58965. Other 
suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et aL, J. 

25 Mol. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 
(Medical Research Council), which are based for example on the ring structure of 
bacterial GroEL or other chaperone polypeptides. 

Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 
30 scaffold and used together with immunoglobulin V H or V L domains to form a ligand. 
Likewise, fibronectin, lipocallin and other scaffolds may be combined. 



WO 2004/003019 PCT/GB2003/002804 

12 

In the case that the variable domains are selected from V~gene repertoires selected for 
instance using phage display technology as herein described, then these variable domains 
can comprise a universal framework region, such that is they may be recognised by a 
specific generic ligand as herein defined. The use of universal frameworks, generic 
5 ligands and the like is described in WO99/20749. In the present invention, reference to 
phage display includes the use of both phage and/or phagemids. 

Where V-gene repertoires are used variation in polypeptide sequence is preferably located 
within the structural loops of the variable domains. The polypeptide sequences of either 
10 variable domain may be altered by DNA shuffling or by mutation in order to enhance the 
interaction of each variable domain with its complementary pair. 

In a preferred embodiment of the invention the 'dual-specific ligand' is a single chain Fv 
fragment. In an alternative embodiment of the invention, the 'dual-specific ligand 5 
15 consists of a Fab region of an antibody. The term "Fab region" includes a Fab-like 
region where two VH or two VL domains are used. 

The variable regions may be derived from antibodies directed against target antigens or 
epitopes. Alternatively they may be derived from a repertoire of single antibody domains 
20 such as those expressed on the surface of filamentous bacteriophage. Selection may be 
performed as described below. 

In a third aspect, the invention provides a method for producing a ligand comprising a 
first immunoglobulin single variable domain having a first binding specificity and a 
25 second single immunoglobulin single variable domain having a second (different) binding 
specificity, one or both of the binding specificities being specific for an antigen which 
increases the half-life of the ligand in vivo, the method comprising the steps of: 

(a) selecting a first variable domain by its ability to bind to a first epitope, 

(b) selecting a second variable region by its ability to bind to a second epitope, 
30 (c) combining the variable domains; and 

(d) selecting the ligand by its ability to bind to said first epitope and to said second 
epitope. 



WO 2004/003019 



PCT/GB2003/002804 



13 

The ligand can bind to the first and second epitopes either simultaneously or, where there 
is competition between the binding domains for epitope binding, the binding of one 
domain may preclude the binding of another domain to its cognate epitope. In one 
embodiment, therefore, step (d) above requires simultaneous binding to both first and 
5 second (and possibly further) epitopes; in another embodiment, the binding to the first 
and second epitoes is not simultaneous. 

The epitopes are preferably on separate antigens. 

10 Ligands advantageously comprise V H /V L combinations, or V H /V H or Vl/V l combinations 
of immunoglobulin variable domains, as described above. The ligands may moreover 
comprise camelid V H h domains, provided that the V H h domain which is specific for an 
antigen which increases the half-life of the ligand in vivo does not bind Hen egg white 
lysozyme (HEL), porcine pancreatic alpha-amylase or NmC-A; hcg, BSA-linked RR6 azo 

15 dye or S. mutans HG982 cells, as described in Conrath et al, (2001) JBC 276:7346-7350 
and W099/23221, neither of which describe the use of a specificity for an antigen which 
increases half-life to increase the half life of the ligand in vivo. 

In one embodiment, said first variable domain is selected for binding to said first epitope 
20 in absence of a complementary variable domain. In a further embodiment, said first 
variable domain is selected for binding to said first epitope/antigen in the presence of a 
third variable domain in which said third variable domain is different from said second 
variable domain and is complementary to the first domain. Similarly, the second domain 
may be selected in the absence or presence of a complementary variable domain. 

25 

The antigens or epitopes targeted by the ligands of the invention, in addition to the half- 
life enhancing protein, may be any antigen or epitope but advantageously is an antigen or 
epitope that is targeted with therapeutic benefit. The invention provides ligands, 
including open conformation, closed conformation and isolated dAb monomer ligands, 
30 specific for any such target, particularly those targets further identified herein. Such 
targets may be, or be part of, polypeptides, proteins or nucleic acids, which may be 
naturally occurring or synthetic. In this respect, the ligand of the invention may bind the 
epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist). One 



WO 2004/003019 PCT/GB2003/002804 

14 

skilled in the art will appreciate that the choice is large and varied. They may be for 
instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for 
enzymes or DNA binding proteins. Suitable cytokines and growth factors include but are 
not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, 
Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, 
FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF-pl, insulin, IFN-y, 
IGF-I, IGF-II, IL-la, IL-ip, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 
a.a.), IL-9, IL-10, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Inhibin a, 
Inhibin p, IP- 10, keratinocyte growth factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, 
Mullerian inhibitory substance, monocyte colony inhibitory factor, monocyte attractant 
protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-1 (MCAF), MCP-2, MCP-3, MCP- 
4, MDC (67 a.a.), MDC (69 a.a.), MIG, MlP-la, MIP-lp, MIP-3a, MIP-3P, MIP-4, 
myeloid progenitor inhibitor factor- 1 (MPIF-1), NAP-2, Neurturin, Nerve growth factor, 
P-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, 
SDFla, SDFlp, SCF, SCGF, stem cell factor (SCF), TARC, TGF-a, TGF-P, TGF-p2, 
TGF-P3, tumour necrosis factor (TNF), TNF-a, TNF-P, TNF receptor I, TNF receptor II, 
TNIL-1, TPO, VEGF, VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, 
GRO/MGSA, GRO-p, GRO-y, HCC1, 1-309, HER 1, HER 2, HER 3 and HER 4. 
Cytokine receptors include receptors for the foregoing cytokines. It will be appreciated 
that this list is by no means exhaustive. 

In one embodiment of the invention, the variable domains are derived from a respective 
antibody directed against the antigen or epitope. In a preferred embodiment the variable 
domains are derived from a repertoire of single variable antibody domains. 

Ih a further aspect, the present invention provides one or more nucleic acid molecules 
encoding at least a dual-specific ligand as herein defined. The dual specific ligand may 
be encoded on a single nucleic acid molecule; alternatively, each domain may be encoded 
by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid 
molecule, the domains may be expressed as a fusion polypeptide, in the manner of a scFv 
molecule, or may be separately expressed and subsequently linked together, for example 
using chemical linking agents. Ligands expressed from separate nucleic acids will be 
linked together by appropriate means. 



WO 2004/003019 PCT/GB2003/002804 

15 

The nucleic acid may further encode a signal sequence for export of the polypeptides 
from a host cell upon expression and may be fused with a surface component of a 
filamentous bacteriophage particle (or other component of a selection display system) 
5 upon expression. 

In a further aspect the present invention provides a vector comprising nucleic acid 
encoding a dual specific ligand according to the present invention. 

10 In a yet further aspect, the present invention provides a host cell transfected with a vector 
encoding a dual specific ligand according to the present invention. 

Expression from such a vector may be configured to produce, for example on the surface 
of a bacteriophage particle, variable domains for selection. This allows selection of 
15 displayed variable regions and thus selection of 'dual- specific ligands' using the method 
of the present invention. 

The present invention further provides a kit comprising at least a dual-specific ligand 
according to the present invention. 

20 

Dual-Specific ligands according to the present invention preferably comprise 
combinations of heavy and light chain domains. For example, the dual specific ligand 
may comprise a Vh domain and a V L domain, which may be linked together in the form 
of an scFv. In addition, the ligands may comprise one or more Ch or Cl domains. For 

25 example, the ligands may comprise a ChI domain, C H 2 or C H 3 domain, and/or a C L 
domain, Cpl, C|u2, C\i3 or Cjj4 domains, or any combination thereof. A hinge region 
domain may also be included. Such combinations of domains may, for example, mimic 
natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or 
F(ab') 2 molecules. Other structures, such as a single arm of an IgG molecule comprising 

30 Vh, Vl, CrI and Cl domains, are envisaged. 

In a preferred embodiment of the invention, the variable regions are selected from single 
domain V gene repertoires. Generally the repertoire of single antibody domains is 



WO 2004/003019 PCT/GB2003/002804 

16 

displayed on the surface of filamentous bacteriophage. In a preferred embodiment each 
single antibody domain is selected by binding of a phage repertoire to antigen. 

In a preferred embodiment of the invention each single variable domain may be selected 
for binding to its target antigen or epitope in the absence of a complementary variable 
region. In an alternative embodiment, the single variable domains may be selected for 
binding to its target antigen or epitope in the presence of a complementary variable 
region. Thus the first single variable domain may be selected in the presence of a third 
complementary variable domain, and the second variable domain may be selected in the 
presence of a fourth complementary variable domain. The complementary third or fourth 
variable domain may be the natural cognate variable domain having the same specificity 
as the single domain being tested, or a non-cognate complementary domain — such as a 
"dummy" variable domain. 

Preferably, the dual specific ligand of the invention comprises only two variable domains 
although several such ligands may be incorporated together into the same protein, for 
example two such ligands can be incorporated into an IgG or a multimeric 
immunoglobulin, such as IgM. Alternatively, in another embodiment a plurality of dual 
specific ligands are combined to form a multimer. For example, two different dual 
specific ligands are combined to create a tetra-specific molecule. 

It will be appreciated by one skilled in the art that the light and heavy variable regions of 
a dual- specific ligand produced according to the method of the present invention may be 
on the same polypeptide chain, or alternatively, on different polypeptide chains. In the 
case that the variable regions are on different polypeptide chains, then they may be linked 
via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking 
group, or any other method known in the art. 

In a further aspect, the present invention provides a composition comprising a dual- 
specific ligand, obtainable by a method of the present invention, and a pharmaceutically 
acceptable carrier, diluent or excipient. 



WO 2004/003019 PCT/GB2003/002804 

17 

Moreover, the present invention provides a method for the treatment and/or prevention 
of disease using a 'dual- specific ligand' or a composition according to the present 
invention. 

5 In a second configuration, the present invention provides multispecific ligands which 
comprise at least two non-complementary variable domains. For example, the ligands 
may comprise a pair of V H domains or a pair of Vl domains. Advantageously, the 
domains are of non-camelid origin; preferably they are human domains or comprise 
human framework regions (FWs) and one or more heterologous CDRs. CDRs and 
10 framework regions are those regions of an immunoglobulin variable domain as defined in 
the Kabat database of Sequences of Proteins of Immunological Interest. 

Preferred human framework regions are those encoded by germline gene segments DP47 
and DPK9. Advantageously, FW1, FW2 and FW3 of a Vh or Vl domain have the 
15 sequence of FW1, FW2 or FWS from DP47 or DPK9. The human frameworks may 
optionally contain mutations, for example up to about 5 amino acid changes or up to 
about 10 amino acid changes collectively in the human frameworks used in the ligands of 
the invention. 

20 The variable domains in the multispecific ligands according to the second configuration 
of the invention may be arranged in an open or a closed conformation; that is, they may 
be arranged such that the variable domains can bind their cognate ligands independently 
and simultaneously, or such that only one of the variable domains may bind its cognate 
ligand at any one time. 

25 

The inventors have realised that under certain structural conditions, non-complementary 
variable domains (for example two light chain variable domains or two heavy chain 
variable domains) may be present in a ligand such that binding of a first epitope to a first 
variable domain inhibits the binding of a second epitope to a second variable domain, 
30 even though such non-complementary domains do not operate together as a cognate pair. 



WO 2004/003019 PCT/GB2003/002804 

18 

Advantageously, the ligand comprises two or more pairs of variable domains; that is, it 
comprises at least four variable domains. Advantageously, the four variable domains 
comprise frameworks of human origin. 

In a preferred embodiment, the human frameworks are identical to those of human 
germline sequences. 

The present inventors consider that such antibodies will be of particular use in ligand 
binding assays for therapeutic and other uses. 

Thus, in a first aspect of the second configuration, the present invention provides a 
method for producing a multispecific ligand comprising the steps of: 

a) selecting a first epitope binding domain by its ability to bind to a first epitope, 

b) selecting a second epitope binding domain by its ability to bind to a second 
epitope, 

c) combining the epitope binding domains; and 

d) selecting the closed conformation multispecific ligand by its ability to bind to said 
first second epitope and said second epitope. 

In a further aspect of the second configuration, the invention provides method for 
preparing a closed conformation multi-specific ligand comprising a first epitope binding 
domain having a first epitope binding specificity and a non-complementary second 
epitope binding domain having a second epitope binding specificity, wherein the first and 
second binding specificities compete for epitope binding such that the closed 
conformation multi-specific ligand may not bind both epitopes simultaneously, said 
method comprising the steps of: 

a) selecting a first epitope binding domain by its ability to bind to a first epitope, 

b) selecting a second epitope binding domain by its ability to bind to a second 
epitope, 

c) combining the epitope binding domains such that the domains are in a closed 
conformation; and 



WO 2004/003019 PCT/GB2003/002804 

19 

d) selecting the closed conformation multispecific ligand by its ability to bind to 
said first second epitope and said second epitope, but not to both said first and 
second epitopes simultaneously. 

Moreover, the invention provides a closed conformation multi-specific ligand comprising 
a first epitope binding domain having a first epitope binding specificity and a non- 
complementary second epitope binding domain having a second epitope binding 
specificity, wherein the first and second binding specificities compete for epitope binding 
such that the closed conformation multi-specific ligand may not bind both epitopes 
simultaneously. 

An alternative embodiment of the above aspect of the of the second configuration of the 
invention optionally comprises a further step (bl) comprising selecting a third or further 
epitope binding domain. In this way the multi-specific ligand produced, whether of open 
or closed conformation, comprises more than two epitope binding specificities. In a 
preferred aspect of the second configuration of the invention, where the multi-specific 
ligand comprises more than two epitope binding domains, at least two of said domains are 
in a closed conformation and compete for binding; other domains may compete for 
binding or may be free to associate independently with their cognate epitope(s). 

According to the present invention the term 'multi-specific ligand 5 refers to a ligand 
which possesses more than one epitope binding specificity as herein defined. 

As herein defined the term 'closed conformation' (multi-specific ligand) means that the 
epitope binding domains of the ligand are attached to or associated with each other, 
optionally by means of a protein skeleton, such that epitope binding by one epitope 
binding domain competes with epitope binding by another epitope binding domain. That 
is, cognate epitopes may be bound by each epitope binding domain individually but not 
simultaneosuly. The closed conformation of the ligand can be achieved using methods 
herein described. 

"Open conformation" means that the epitope binding domains of the ligand are attached 
to or associated with each other, optionally by means of a protein skeleton, such that 



WO 2004/003019 



PCT/GB2003/002804 



20 

epitope binding by one epitope binding domain does not compete with epitope binding 
by another epitope binding domain. 

As referred to herein, the term 'competes' means that the binding of a first epitope to its 
5 cognate epitope binding domain is inhibited when a second epitope is bound to its 
cognate epitope binding domain. For example, binding may be inhibited sterically, for 
example by physical blocking of a binding domain or by alteration of the structure or 
environment of a binding domain such that its affinity or avidity for an epitope is reduced. 

10 In a further embodiment of the second configuration of the invention, the epitopes may 
displace each other on binding. For example, a first epitope may be present on an antigen 
which, on binding to its cognate first binding domain, causes steric hindrance of a second 
binding domain, or a coformational change therein, which displaces the epitope bound to 
the second binding domain. 

15 

Advantageously, binding is reduced by 25% or more, advantageously 40%, 50%, 60%, 
70%o, 80%>, 90% or more, and preferably up to 100% or nearly so, such that binding is 
completely inhibited. Binding of epitopes can be measured by conventional antigen 
binding assays, such as ELIS A, by fluorescence based techniques, including FRET, or by 
20 techniques such as suface plasmon resonance which measure the mass of molecules. 

According to the method of the present invention, advantageously, each epitope binding 
domain is of a different epitope binding specificity. 

25 In the context of the present invention, first and second "epitopes" are understood to be 
epitopes which are not the same and are not bound by a single monospecific ligand. They 
may be on different antigens or on the same antigen, but separated by a sufficient distance 
that they do not form a single entity that could be bound by a single mono-specific V H /V L 
binding pair of a conventional antibody. Experimentally, if both of the individual 

30 variable domains in single chain antibody form (domain antibodies or dAbs) are 
separately competed by a monospecific V H /V L ligand against two epitopes then those two 
epitopes are not sufficiently far apart to be considered separate epitopes according to the 
present invention. 



WO 2004/003019 



PCT/GB2003/002804 



21 

The closed conformation multispecific ligands of the invention do not include ligands as 
described in WO 02/02773. Thus, the ligands of the present invention do not comprise 
complementary Vj/Vl pairs which bind any one or more antigens or epitopes co- 
5 operatively. Instead, the ligands according to the invention preferably comprise non- 
complementary V H -V H or V L -V L P airs - Advantageously, each v H or V L domain in each 
Vh~Vh or Vl~Vl P a ^ r has a different epitope binding specificity, and the epitope binding 
sites are so arranged that the binding of an epitope at one site competes with the binding 
of an epitope at another site. 

10 

According to the present invention, advantageously, each epitope binding domain 
comprises an immunoglobulin variable domain. More advantageously, each 
immunoglobulin variable domain will be either a variable light chain domain (Vl) or a 
variable heavy chain domain Vh- I* 1 the second configuration of the present invention, 
15 the immunoglobulin domains when present on a ligand according to the present 
invention are non-complementary, that is they do not associate to form a Vp/Vl antigen 
binding site. Thus, multi-specific ligands as defined in the second configuration of the 
invention comprise immunoglobulin domains of the same sub-type, that is either variable 
light chain domains (Vl) or variable heavy chain domains (Vh)- Moreover, where the 
ligand according to the invention is in the closed conformation, the immunoglobulin 
domains may be of the camelid Vhh type. 

In an alternative embodiment, the ligand(s) according to the invention do not comprise a 
camelid Vhh domain. More particularly, the ligand(s) of the invention do not comprise 
one or more amino acid residues that are specific to camelid Vhh domains as compared to 
human Vh domains. 

Advantageously, the single variable domains are derived from antibodies selected for 
binding activity against different antigens or epitopes. For example, the variable domains 
may be isolated at least in part by human immunisation. Alternative methods are known 
in the art, including isolation from human antibody libraries and synthesis of artificial 
antibody genes. 



WO 2004/003019 PCT/GB2003/002804 

22 

The variable domains advantageously bind superantigens, such as protein A or protein L. 
Binding to superantigens is a property of correctly folded antibody variable domains, and 
allows such domains to be isolated from, for example, libraries of recombinant or mutant 
domains. 

Epitope binding domains according to the present invention comprise a protein scaffold 
and epitope interaction sites (which are advantageously on the surface of the protein 
scaffold). 

Epitope binding domains may also be based on protein scaffolds or skeletons other than 
immunoglobulin domains. For example natural bacterial receptors such as SpA have been 
used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to 
one or more epitopes. Details of this procedure are described in US 5,831,012. Other 
suitable scaffolds include those based on fibronectin and affibodies. Details of suitable 
procedures are described in WO 98/58965. Other suitable scaffolds include lipocallin and 
CTLA4, as described in van den Beuken et al 9 J. Mol. Biol. (2001) 310, 591-601, and 
scaffolds such as those described in WO0069907 (Medical Research Council), which are 
based for example on the ring structure of bacterial GroEL or other chaperone 
polypeptides. 

Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 
scaffold and used together with immuno globulin V H or V L domains to form a multivalent 
ligand. Likewise, fibronectin, lipocallin and other scaffolds may be combined. 

25 It will be appreciated by one skilled in the art that the epitope binding domains of a closed 
conformation multispecific ligand produced according to the method of the present 
invention may be on the same polypeptide chain, or alternatively, on different polypeptide 
chains. In the case that the variable regions are on different polypeptide chains, then they 
may be linked via a linker, advantageously a flexible linker (such as a polypeptide chain), 

30 a chemical linking group, or any other method known in the art. 



10 



15 



20 



WO 2004/003019 PCT/GB2003/002804 

23 

The first and the second epitope binding domains may be associated either covalently or 
non-covalently. In the case that the domains are covalently associated, then the 
association may be mediated for example by disulphide bonds. 

5 In the second configuation of the invention, the first and the second epitopes are 
preferably different. They may be, or be part of, polypeptides, proteins or nucleic acids, 
which may be naturally occurring or synthetic. In this respect, the ligand of the invention 
may bind an epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor 
agonist). The epitope binding domains of the ligand in one embodiment have the same 

10 epitope specificity, and may for example simultaneously bind their epitope when multiple 
copies of the epitope are present on the same antigen. In another embodiment, these 
epitopes are provided on different antigens such that the ligand can bind the epitopes and 
bridge the antigens. One skilled in the art will appreciate that the choice of epitopes and 
antigens is large and varied. They may be for instance human or animal proteins, 

15 cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins. 
Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, 
BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, 
EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine 
(CX3C), GDNF, G-CSF, GM-CSF, GF-(31, insulin, IFN-y, IGF-I, IGF-II, IL-la, IL-ip, 

20 IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL-12, 
IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Miibin a, Inhibin p, IP-10, keratinocyte growth 
factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, Mullerian inhibitory substance, 
monocyte colony inhibitory factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), 
MDC (69 a.a.), MCP-1 (MCAF), MCP-2, MCP-3, MCP-4, MDC (67 a.a.), MDC (69 

25 a.a.), MIG, MlP-la, MIP-ip, MIP-3a, MIP-3P, MIP-4, myeloid progenitor inhibitor 
factor-1 (MPIF-1), NAP-2, Neurturin, Nerve growth factor, P-NGF, NT-3, NT-4, 
Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFloc, SDFip, SCF, 
SCGF, stem cell factor (SCF), TARC, TGF-a, TGF-p, TGF-P2, TGF-p3, tumour necrosis 
factor (TNF), TNF-a, TNF-p, TNF receptor I, TNF receptor II, TNIL-1, TPO, VEGF, 

30 VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, GRO/MGSA, GRO-p, 
GRO-y, HCC1, 1-309, HER 1, HER 2, HER 3, HER 4, TACE recognition site, TNF BP-I 
and TNF BP-II, as well as any target disclosed in Annex 2 or Annex 3 hereto, whether in 
combination as set forth in the Annexes, in a different combination or individually. 



WO 2004/003019 PCT/GB2003/002804 

24 

Cytokine receptors include receptors for the foregoing cytokines, e.g. IL-1 Rl; IL-6R; 
IL-10R; IL-18R, as well as receptors for cytokines set forth in Annex 2 or Annex 3 and 
also receptors disclosed in Annex 2 and 3. It will be appreciated that this list is by no 
means exhaustive. Where the multispecific ligand binds to two epitopes (on the same or 
5 different antigens), the antigen(s) may be selected from this list. 

Advantageously, dual specific ligands may be used to target cytokines and other 
molecules which cooperate synergistically in therapeutic situations in the body of an 
organism. The invention therefore provides a method for synergising the activity of two 

10 or more cytokines, comprising administering a dual specific ligand capable of binding to 
said two or more cytokines. In this aspect of the invention, the dual specific ligand may 
be any dual specific ligand, including a ligand composed of complementary and/or non- • 
complementary domains, a ligand in an open conformation, and a ligand in a closed 
conformation. For example, this aspect of the invention relates to combinations of V H 

15 domains and V L domains, V H domains only and V L domains only. 

Synergy in a therapeutic context may be achieved in a number of ways. For example, 
target combinations may be therapeutically active only if both targets are targeted by the 
ligand, whereas targeting one target alone is not therapeutically effective. In another 
20 embodiment, one target alone may provide some low or minimal therapeutic effect, but 
together with a second target the combination provides a synergistic increase in 
therapeutic effect. 

Preferably, the cytokines bound by the dual specific ligands of this aspect of the invention 
25 are sleeted from the list shown in Annex 2. 

Moreover, dual specific ligands may be used in oncology applications, where one 
specificity targets CD89, which is expressed by cytotoxic cells, and the other is tumour 
specific. Examples of tumour antigens which may be targetted are given in Annex 3. 

30 

In one embodiment of the second configuration of the invention, the variable domains are 
derived from an antibody directed against the first and/or second antigen or epitope. In a 
preferred embodiment the variable domains are derived from a repertoire of single 



WO 2004/003019 



PCT/GB2003/002804 



25 

variable antibody domains. In one example, the repertoire is a repertoire that is not 
created in an animal or a synthetic repertoire. In another example, the single variable 
domains are not isolated (at least in part) by animal immunisation. Thus, the single 
domains can be isolated from a naive library. 

The second configuration of the invention, in another aspect, provides a multi-specific 
ligand comprising a first epitope binding domain having a first epitope binding specificity 
and a non-complementary second epitope binding domain having a second epitope 
binding specificity. The first and second binding specificities may be the same or 
different. 

In a further aspect, the present invention provides a closed conformation multi-specific 
ligand comprising a first epitope binding domain having a first epitope binding specificity 
and a non-complementary second epitope binding domain having a second epitope 
binding specificity wherein the first and second binding specificities are capable of 
competing for epitope binding such that the closed conformation multi-specific ligand 
cannot bind both epitopes simultaneously. 

In a still further aspect, the invention provides open conformation ligands comprising 
non-complementary binding domains, wherein the deomains are specific for a different 
epitope on the same target. Such ligands bind to targets with increased avidity. 
Similarly, the invention provides multivalent ligands comprising non-complementary 
binding domains specific for the same epitope and directed to targets which comprise 
multiple copies of said epitope, such as IL-5, PDGF-AA, PDGF-BB, TGF beta, TGF 
beta2, TGF beta3 and TNFa, for eample human TNF Receptor 1 and human TNFoe. 

In a similar aspect, ligands according to the invention can be configured to bind 
individual epitopes with low affinity, such that binding to individual epitopes is not 
therapeutically significant; but the increased avidity resulting from binding to two 
epitopes provides a theapeutic benefit. In a perticular example, epitopes may be targetted 
which are present individually on normal cell types, but present together only on 
abnormal or diseased cells, such as tumour cells. In such a situaton, only the abnormal or 



WO 2004/003019 PCT/GB2003/002804 

26 

diseased cells are effectively targetted by the bispecific ligands according to the 
invention. 

Ligand specific for multiple copies of the same epitope, or adjacent epitopes, on the same 
5 target (known as chelating dAbs) may also be trimeric or polymeric (tertrameric or more) 
ligands comprising three, four or more non-complementary binding domains. For 
example, ligands may be constructed comprising three or four V H domains or V L 
domains. 

10 Moreover, ligands are provided which bind to multisubunit targets, wherein each binding 
domain is specific for a subunit of said target. The ligand may be dimeric, trimeric or 
polymeric. 

Preferably, the multi-specific ligands according to the above aspects of the invention are 
15 obtainable by the method of the first aspect of the invention. 

According to the above aspect of the second configuration of the invention, 
advantageously the first epitope binding domain and the second epitope binding domains 
are non- complementary immunoglobulin variable domains, as herein defined. That is 
20 either Vh"V h or V L ~V L variable domains. 

Chelating dAbs in particular may be prepared according to a preferred aspect of the 
invention, namely the use of anchor dAbs, in which a library of dimeric, trimeric or 
multimeric dAbs is constructed using a vector which comprises a constant dAb upstream 
25 or downstream of a linker sequence, with a repertoire of second, third and further dAbs 
being inserted on the other side of the linker. For example, the anchor or guiding dAb 
may be TAR1-5 (Vk), TAR1-27(Vk), TAR2h-5(VH) or TAR2h-6(Vic). 

In alternative methodologies, the use of linkers may be avoided, for example by the use of 
30 non-covalent bonding or naturall affinity between binding domains such as V H and V K . 
The invention accordingly provides a method for preparing a chelating multimeric ligand 
comprising the steps of: 



WO 2004/003019 PCT/GB2003/002804 

27 

(a) providing a vector comprising a nucleic acid sequence encoding a single 
binding domain specific for a first epitope on a target; 

(b) providing a vector encoding a repertoire comprising second binding domains 
specific for a second epitope on said target, which epitope can be the same or different to 

5 the first epitope, said second epitope being adjacent to said first epitope; and 

(c) expressing said first and second binding domains; and 

(d) isolating those combinations of first and second binding domains which 
combine together to produce a target-binding dimer. 

10 The first and second epitopes are adjacent such that a multimeric ligand is capable of 
binding to both epitopes simultaneously. This provides the ligand with the advantages of 
increased avidity if binding. Where the epitopes are the same, the increased avidity is 
obtained by the presence of multiple copies of the epitope on the target, allowing at least 
two copies to be simultaneously bound in order to obtain the increased avidity effect. 

15 

The binding domains may be associated by several methods, as well as the use of linkers. 
For example, the binding domains may comprise cys residues, avidin and streptavidin 
groups or other means for non-covalent attachment post-synthesis; those combinations 
which bind to the target efficiently will be isolated. Alternatively, a linker may be present 
20 between the first and second binding domains, which are expressed as a single 
polypeptide from a single vector, which comprises the first binding domain, the linker 
and a repertoire of second binding domains, for instance as described above. 

In a preferred aspect, the first and second binding domains associate naturally when 
25 bound to antigen; for example, V H and V K domains, when bound to adjacent epitopes, will 
naturally associate in a three-way interaction to form a stable dimer. Such associated 
proteins can be isolated in a target binding assay. An advantage of this procedure is that 
only binding domains which bind to closely adjacent epitopes, in the correct 
conformation, will associate and thus be isolated as a result of their increased avidity for 
30 the target. 

In an alternative embodiment of the above aspect of the second configuration of the 
invention, at least one epitope binding domain comprises a non-immuno globulin 'protein 



WO 2004/003019 



PCT/GB2003/002804 



28 

scaffold' or 'protein skeleton' as herein defined. Suitable non-immunoglobulin protein 
scaffolds include but are not limited to any of those selected from the group consisting of: 
SpA, fibronectin, GroEL and other chaperones, lipocallin, CCTLA4 and affibodies, as set 
forth above. 

According to the above aspect of the second configuration of the invention, 
advantageously, the epitope binding domains are attached to a 'protein skeleton'. 
Advantageously, a protein skeleton according to the invention is an immunoglobulin 
skeleton. 

According to the present invention, the term 'immunoglobulin skeleton' refers to a 
protein which comprises at least one immunoglobulin fold and which acts as a nucleus for 
one or more epitope binding domains, as defined herein. 

Preferred immunoglobulin skeletons as herein defined includes any one or more of those 
selected from the following: an immunoglobulin molecule comprising at least (i) the CL 
(kappa or lambda subclass) domain of an antibody; or (ii) the CHI domain of an antibody 
heavy chain; an immunoglobulin molecule comprising the CHI and CH2 domains of an 
antibody heavy chain; an immunoglobulin molecule comprising the CHI, CH2 and CHS 
domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL 
(kappa or lambda subclass) domain of an antibody. A hinge region domain may also be 
included. Such combinations of domains may, for example, mimic natural antibodies, 
such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules. 
Those skilled in the art will be aware that this list is not intended to be exhaustive. 

Linking of the skeleton to the epitope binding domains, as herein defined may be 
achieved at the polypeptide level, that is after expression of the nucleic acid encoding the 
skeleton and/or the epitope binding domains. Alternatively, the linking step may be 
performed at the nucleic acid level. Methods of linking a protein skeleton according to the 
present invention, to the one or more epitope binding domains include the use of protein 
chemistry and/or molecular biology techniques which will be familiar to those skilled in 
the art and are described herein. 



WO 2004/003019 PCT/GB2003/002804 

29 

Advantageously, the closed conformation multispecific ligand may comprise a first 
domain capable of binding a target molecule, and a second domain capable of binding a 
molecule or group which extends the half-life of the ligand. For example, the molecule or 
group may be a bulky agent, such as HSA or a cell matrix protein. As used herein, the 
5 phrase "molecule or group which extends the half-life of a ligand" refers to a molecule or 
chemical group which, when bound by a dual-specific ligand as described herein 
increases the in vivo half-life of such dual specific ligand when administered to an 
animal, relative to a ligand that does not bind that molecule or group. Examples of 
molecules or groups that extend the half-life of a ligand are described hereinbelow. In a 

10 preferred embodiment, the closed conformation multispecific ligand may be capable of 
binding the target molecule only on displacement of the half-life enhancing molecule or 
group. Thus, for example, a closed conformation multispecific ligand is maintained in 
circulation in the bloodstream of a subject by a bulky molecule such as HSA. When a 
target molecule is encountered, competition between the binding domains of the closed 

15 conformation multispecific ligand results in displacement of the HSA and binding of the 
target. 

Ligands according to any aspect of the present invention, as well as dAb monomers 
useful in constructing such ligands, may advantageously dissociate from their cognate 

20 target(s) with a Kd of 300nM to 5pM (ie, 3 x 10"" 7 to 5 x 10~ 12 M), preferably 50nM 
to20pM, or 5nM to 200pM or InM to lOOpM, 1 x 1(T 7 M or less, 1 x 10" 8 M or less, 1 x 
10~ 9 M or less, 1 x 10" 10 M or less, 1 x 10" 11 M or less; and/or a Koff rate constant of 5 x 
10' 1 to 1 x 10" 7 S'\ preferably 1 x 10" 2 to 1 x 10~ 6 S" 1 , or 5 x 10' 3 to 1 x 10* 5 S'\ or 5 x 10" 1 
S" 1 or less, or 1 x 10" 2 S" 1 or less, or 1 x 10* 3 S" 1 or less, or 1 x 10~ 4 S" 1 or less, or 1 x 10" 5 S~ l 

25 or less, or 1 x 1CT 6 S" 1 or less as determined by surface plasmon resonance. The Kd rate 
constand is defined as Kcff/K^. 

In particular the invention provides an anti-TNFa dAb monomer (or dual specific ligand 
comprising such a dAb), homodimer, heterodimer or homotrimer ligand, wherein each 
30 dAb binds TNFoc. The ligand binds to TNFcc with a Kd of 300nM to 5pM (ie, 3 x 1(T 7 to 
5 x 10" 12 M), preferably 50nM to 20pM, more preferably 5nM to 200pM and most 
preferably InM to lOOpM; expressed in an alternative manner, the Kd is 1 x 10" 7 M or 
less, preferably 1x10" M or less, more preferably 1 x 10 M or less, advantageously 1 x 



WO 2004/003019 PCT/GB2003/002804 

30 

10" 10 M or less and most preferably 1 x 10' 11 M or less; and/or a Koff rate constant of 5 x 
10" 1 to 1 x 10" 7 S~\ preferably 1 x 10~ 2 to 1 x 10~ 6 S" 1 , more preferably 5 x 10~ 3 to 1 x 10" 5 
S" 1 , for example 5 x 10" 1 S" 1 or less, preferably 1 x 10" 2 S" 1 or less, more preferably 1x10" 
3 S" 1 or less, advantageously 1 x 10~ 4 S" 1 or less, further advantageously 1 x 10" 5 S* 1 or less, 
5 and most preferably 1x10" S" or less, as determined by surface plasmon resonance. 

Preferably, the ligand neutralises TNFoc in a standard L929 assay with an ND50 of 
500nM to 50pM, preferably or lOOnM to 50pM, advantageously lOnM to lOOpM, more 
preferably InM to lOOpM; for example 50nM or less, preferably 5nM or less, 
10 advantageously 500pM or less, more preferably 200pM or less and most preferably 
lOOpM or less. 

Preferably, the ligand inhibits binding of TNF alpha to TNF alpha Receptor I (p55 
receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably 
15 lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 
5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most 
preferably lOOpM or less. Preferably, the TNFoc is Human TNF a. 

Furthermore, the invention provides a an anti-TNF Receptor I dAb monomer, or dual 
20 specific ligand comprising such a dAb, that binds to TNF Receptor I with a IQ of 300nM 
to 5pM (ie, 3 x 10' 7 to 5 x 10' 12 M), preferably 50nM to20pM, more preferably 5nM to 
200pM and most preferably InM to lOOpM, for example 1 x 10" 7 M or less, preferably 1 
x 10" 8 M or less, more preferably 1 x 10" 9 M or less, advantageously 1 x 10" 10 M or less 
and most preferably 1 x 10" 11 M or less; and/or a Koff rate constant of 5 x 10" 1 to 1 x 10" 7 
25 S'\ preferably 1 x 10" 2 to 1 x 10" 6 S"\ more preferably 5 x 10" 3 to 1 x 10' 5 S" 1 , for 
example 5 x 10" 1 S" 1 or less, preferably 1 x 10" 2 S" 1 or less, advantageously 1 x 10" 3 S" 1 or 
less, more preferably 1 x lO^S" 1 or less, still more preferably 1 x 10" 5 S" 1 or less, and most 
preferably 1x10 S or less as determined by surface plasmon resonance. 

30 Preferably, the dAb monomeror ligand neutralises TNFoc in a standard assay (eg, the 
L929 or HeLa assays described herein) with an ND50 of 500nM to 50pM, preferably 
lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for 



WO 2004/003019 PCT/GB2003/002804 

31 

example 50nM or less, preferably 5nM or less, more preferably 500pM or less, 
advantageously 200pM or less, and most preferably lOOpM or less. 

Preferably, the dAb monomer or ligand inhibits binding of TNF alpha to TNF alpha 
5 Receptor I (p55 receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, 
more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or 
less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or 
less, and most preferably lOOpM or less. Preferably, the TNF Receptor I target is Human 
TNFcc. 

10 

Furthermore, the invention provides a dAb monomer(or dual specific ligand comprising 
such a dAb) that binds to serum albumin (SA) with a Kid of InM to 500/xM (ie, x 10~ 9 to 
5x10"), preferably lOOnM to 10/xM. Preferably, for a dual specific ligand comprising a 
first anti-SA dAb and a second dAb to another target, the affinity (eg and/or Ko ff as 

15 measured by surface plasmon resonance, eg using BiaCore) of the second dAb for its 
target is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 
100000, or 10000 to 100000 times) the affinity of the first dAb for SA. For example, the 
first dAb binds SA with an affinity of approximately 10/xM, while the second dAb binds 
its target with an affinity of lOOpM. Preferably, the serum albumin is human serum 

20 albumin (HSA). 

In one embodiment, the first dAb (or a dAb monomer) binds SA (eg, HSA) with a K4 of 
approximately 50, preferably 70, and more preferably 100, 150 or 200 nM. 

25 The invention moreover provides dimers, trimers and polymers of the aforementioned 
dAb monomers, in accordance with the foregoing aspect of the present invention. 

Ligands according to the invention, including dAb monomers, dimers and trimers, can be 
linked to an antibody Fc region, comprising one or both of Cr2 and Ch3 domains, and 
30 optionally a hinge region. For example, vectors encoding ligands linked as a single 
nucleotide sequence to an Fc region may be used to prepare such polypeptides. 



WO 2004/003019 PCT/GB2003/002804 

32 

In a further aspect of the second configuration of the invention, the present invention 
provides one or more nucleic acid molecules encoding at least a multispecific ligand as 
herein defined. In one embodiment, the ligand is a closed conformation ligand. In 
another embodiment, it is an open conformation ligand. The multispecific ligand may be 
5 encoded on a single nucleic acid molecule; alternatively, each epitope binding domain 
may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a 
single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, or 
may be separately expressed and subsequently linked together, for example using 
chemical linking agents. Ligands expressed from separate nucleic acids will be linked 
10 together by appropriate means. 

The nucleic acid may further encode a signal sequence for export of the polypeptides 
from a host cell upon expression and may be fused with a surface component of a 
filamentous bacteriophage particle (or other component of a selection display system) 
15 upon expression. Leader sequences, which may be used in bacterial expresion and/or 
phage or phagemid display, include pelB, stll, ompA, phoA, bla and pelA. 

In a further aspect of the second configuration of the invention the present invention 
provides a vector comprising nucleic acid according to the present invention. 

20 

In a yet further aspect, the present invention provides a host cell transfected with a vector 
according to the present invention. 

Expression from such a vector may be configured to produce, for example on the surface 
25 of a bacteriophage particle, epitope binding domains for selection. This allows selection 
of displayed domains and thus selection of 'multispecific ligands' using the method of the 
present invention. 

In a preferred embodiment of the second configuration of the invention, the epitope 
30 binding domains are immunoglobulin variable regions and are selected from single 
domain V gene repertoires. Generally the repertoire of single antibody domains is 
displayed on the surface of filamentous bacteriophage. In a preferred embodiment each 
single antibody domain is selected by binding of a phage repertoire to antigen. 



WO 2004/003019 



PCT/GB2003/002804 



33 

The present invention further provides a kit comprising at least a multispecific ligand 
according to the present invention, which may be an open conformation or closed 
conformation ligand. Kits according to the invention may be, for example, diagnostic 
kits, therapeutic kits, kits for the detection of chemical or biological species, and the like. 

In a further aspect still of the second configuration of the invention, the present invention 
provides a homogenous immunoassay using a ligand according to the present invention. 

In a further aspect still of the second configuration of the invention, the present invention 
provides a composition comprising a closed conformation multispecific ligand, obtainable 
by a method of the present invention, and a pharmaceutically acceptable carrier, diluent or 
excipient. 

Moreover, the present invention provides a method for the treatment of disease using a 
"closed conformation multispecific ligand 5 or a composition according to the present 
invention. 

In a preferred embodiment of the invention the disease is cancer or an inflammatory 
disease, eg rheumatoid arthritis, asthma or Crohn's disease. 

In a further aspect of the second configuration of the invention, the present invention 
provides a method for the diagnosis, including diagnosis of disease using a closed 
conformation multispecific ligand, or a composition according to the present invention. 
Thus in general the binding of an analyte to a closed conformation multispecific ligand 
may be exploited to displace an agent, which leads to the generation of a signal on 
displacement. For example, binding of analyte (second antigen) could displace an 
enzyme (first antigen) bound to the antibody providing the basis for an immunoassay, 
especially if the enzyme were held to the antibody through its active site. 

Thus in a final aspect of the second configuration, the present invention provides a 
method for detecting the presence of a target molecule, comprising: 



WO 2004/003019 



PCT/GB2003/002804 



34 

(a) providing a closed conformation multispecific ligand bound to an agent, said ligand 
being specific for the target molecule and the agent, wherein the agent which is bound by 
the ligand leads to the generation of a detectable signal on displacement from the ligand; 

(b) exposing the closed conformation multispecific ligand to the target molecule; and 

(c) detecting the signal generated as a result of the displacement of the agent. 

According to the above aspect of the second configuration of the invention, 
advantageously, the agent is an enzyme, which is inactive when bound by the closed 
conformation multi-specific ligand. Alternatively, the agent may be any one or more 
selected from the group consisting of the following: the substrate for an enzyme, and a 
fluorescent, luminescent or chromogenic molecule which is inactive or quenched when 
bound by the ligand. 

Sequences similar or homologous (e.g., at least about 70% sequence identity) to the 
sequences disclosed herein are also part of the invention. In some embodiments, the 
sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 
94%, 95%, 96%, 97%, 98%, 99% or higher. At the nucleic acid level, the sequence 
identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 
97%, 98%, 99% or higher. Alternatively, substantial identity exists when the nucleic acid 
segments will hybridize under selective hybridization conditions (e.g., very high 
stringency hybridization conditions), to the complement of the strand. The nucleic acids 
may be present in whole cells, in a cell lysate, or in a partially purified or substantially 
pure form. 

Calculations of "homology" or "sequence identity" or "similarity" between two 
sequences (the terms are used interchangeably herein) are performed as follows. The 
sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in 
one or both of a first and a second amino acid or nucleic acid sequence for optimal 
alignment and non-homologous sequences can be disregarded for comparison purposes). 
In a preferred embodiment, the length of a reference sequence aligned for comparison 
purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more 
preferably at least 60%, and even more preferably at least 70%, 80%), 90%, 100% of the 
length of the reference sequence. The amino acid residues or nucleotides at 



WO 2004/003019 



PCT/GB2003/002804 



35 

corresponding amino acid positions or nucleotide positions are then compared. When a 
position in the first sequence is occupied by the same amino acid residue or nucleotide as 
the corresponding position in the second sequence, then the molecules are identical at that 
position (as used herein amino acid or nucleic acid "homology" is equivalent to amino 
acid or nucleic acid "identity"). The percent identity between the two sequences is a 
function of the number of identical positions shared by the sequences, taking into account 
the number of gaps, and the length of each gap, which need to be introduced for optimal 
alignment of the two sequences. 

Advantageously, the BLAST algorithm (version 2.0) is employed for sequence alignment, 
with parameters set to default values. The BLAST algorithm is described in detail at the 
world wide web site ("www") of the National Center for Biotechnology Information 
(".ncbi") of the National Institutes of Health ("nih") of the U.S. government (".gov"), in 
the "/Blast/" directory, in the "blast_help.html" file. The search parameters are defined as 
follows, and are advantageously set to the defined default parameters. 

BLAST (Basic Local Alignment Search Tool) is the heuristic search algorithm employed 
by the programs blastp, blastn, blastx, tblastn, and tblastx; these programs ascribe 
significance to their findings using the statistical methods of Karlin and Altschul, 1990, 
Proc. Natl. Acad. Sci. USA 87(6):2264-8 (see the "blast_help.html" file, as described 
above) with a few enhancements. The BLAST programs were tailored for sequence 
similarity searching, for example to identify homologues to a query sequence. The 
programs are not generally useful for motif-style searching. For a discussion of basic 
issues in similarity searching of sequence databases, see Altschul et al. (1994). 

The five BLAST programs available at the National Center for Biotechnology 
Information web site perform the following tasks: 

"blastp" compares an amino acid query sequence against a protein sequence database; 
"blastn" compares a nucleotide query sequence against a nucleotide sequence database; 
"blastx" compares the six-frame conceptual translation products of a nucleotide query 
sequence (both strands) against a protein sequence database; 

"tblastn" compares a protein query sequence against a nucleotide sequence database 
dynamically translated in all six reading frames (both strands). 



WO 2004/003019 



PCT/GB2003/002804 



36 

"tblastx" compares the six-frame translations of a nucleotide query sequence against the 
six-frame translations of a nucleotide sequence database. 
BLAST uses the following search parameters: 

HISTOGRAM Display a histogram of scores for each search; default is yes. (See 
5 parameter H in the BLAST Manual). 

DESCRIPTIONS Restricts the number of short descriptions of matching sequences 
reported to the number specified; default limit is 100 descriptions. (See parameter V in 
the manual page). See also EXPECT and CUTOFF. 

ALIGNMENTS Restricts database sequences to the number specified for which high- 
10 scoring segment pairs (HSPs) are reported; the default limit is 50. If more database 
sequences than this happen to satisfy the statistical significance threshold for reporting 
(see EXPECT and CUTOFF below), only the matches ascribed the greatest statistical 
significance are reported. (See parameter B in the BLAST Manual). 

EXPECT The statistical significance threshold for reporting matches against database 
15 sequences; the default value is 10, such that 10 matches are expected to be found merely 
by chance, according to the stochastic model of Karlin and Altschul (1990). If the 
statistical significance ascribed to a match is greater than the EXPECT threshold, the 
match will not be reported. Lower EXPECT thresholds are more stringent, leading to 
fewer chance matches being reported. Fractional values are acceptable. (See parameter E 
20 in the BLAST Manual). 

CUTOFF Cutoff score for reporting high-scoring segment pairs. The default value is 
calculated from the EXPECT value (see above). HSPs are reported for a database 
sequence only if the statistical significance ascribed to them is at least as high as would be 
ascribed to a lone HSP having a score equal to the CUTOFF value. Higher CUTOFF 
25 values are more stringent, leading to fewer chance matches being reported. (See 
parameter S in the BLAST Manual). Typically, significance thresholds can be more 
intuitively managed using EXPECT. 

MATRIX Specify an alternate scoring matrix for BLASTP, BLASTX, TBLASTN and 
TBLASTX. The default matrix is BLOSUM62 (Henikoff & Henikoff, 1992, Proc. Natl. 
30 Aacad. Sci. USA 89(22): 10915-9). The valid alternative choices include: PAM40, 
PAM120, PAM250 and IDENTITY. No alternate scoring matrices are available for 
BLASTN; specifying the MATRIX directive in BLASTN requests returns an error 
response. 



WO 2004/003019 



PCT/GB2003/002804 



37 

STRAND Restrict a TBLASTN search to just the top or bottom strand of the database 
sequences; or restrict a BLASTN, BLASTX or TBLASTX search to just reading frames 
on the top or bottom strand of the query sequence. 

FILTER Mask off segments of the query sequence that have low compositional 
5 complexity, as determined by the SEG program of Wootton & Federhen (1993) 
Computers and Chemistry 17:149-163, or segments consisting of short-periodicity 
internal repeats, as determined by the XNU program of Claverie & States, 1993, 
Computers and Chemistry 17:191-201, or, for BLASTN, by the DUST program of 
Tatusov and Lipman (see the world wide web site of the NCBI). Filtering can eliminate 
10 statistically significant but biologically uninteresting reports from the blast output (e.g., 
hits against common acidic-, basic- or pro line-rich regions), leaving the more biologically 
interesting regions of the query sequence available for specific matching against database 
sequences. 

15 Low complexity sequence found by a filter program is substituted using the letter "N" in 
nucleotide sequence (e.g., "N" repeated 13 times) and the letter "X" in protein sequences 
(e.g., "X" repeated 9 times). 

Filtering is only applied to the query sequence (or its translation products), not to 
20 database sequences. Default filtering is DUST for BLASTN, SEG for other programs. 

It is not unusual for nothing at all to be masked by SEG, XNU, or both, when applied to 
sequences in SWISS-PROT, so filtering should not be expected to always yield an effect. 
Furthermore, in some cases, sequences are masked in their entirety, indicating that the 
statistical significance of any matches reported against the unfiltered query sequence 
25 should be suspect. 

NCBI-gi Causes NCBI gi identifiers to be shown in the output, in addition to the 
accession and/or locus name. 

30 Most preferably, sequence comparisons are conducted using the simple BLAST search 
algorithm provided at the NCBI world wide web site described above, in the "/BLAST" 
directory. 



WO 2004/003019 



PCT/GB2003/002804 



38 

Brief Description of the Figures 

Figure 1 shows the diversification of V H /HSA at positions H50, H52, H52a, H53, 

H55, H56, H58, H95, H96, H97, H98 (DVT or NNK encoded 
respectively) which are in the antigen binding site of V H HSA. The 
sequence of V K is diversified at positions L50, L53. 



Figure 2 shows Library 1 : Germline V K /DVT Vh, 

Library 2: Germline V K /NNK V H , 

Library 3: Germline V H /DVT V K 

Library 4: Germline V H /NNK V K 
In phage display/ScFv format. These libraries were pre-selected for 
binding to generic ligands protein A and protein L so that the majority of 
the clones and selected libraries are functional. Libraries were selected on 
HSA (first round) and /3-gal (second round) or HSA /3-gal selection or on 
/?-gal (first round) and HSA (second round) /3-gal HSA selection. Soluble 
scFv from these clones of PCR are amplified in the sequence. One clone 
encoding a dual specific antibody K8 was chosen for further work. 



Figure 3 shows an alignment of Vh chains and V K chains. 



Figure 4 shows the characterisation of the binding properties of the K8 antibody, 

the binding properties of the K8 antibody characterised by monoclonal 
phage ELISA, the dual specific K8 antibody was found to bind HSA and 
jS-gal and displayed on the surface of the phage with absorbant signals 
greater than 1.0. No cross reactivity with other proteins was detected. 



Figure 5 shows soluble scFv ELISA performed using known concentrations of the 

K8 antibody fragment. A 96-well plate was coated with 100/ig of HSA, 
BSA and /3-gal at lOjttg/ml and 100/xg/ml of Protein A at 1/ig/ml 
concentration. 50 fig of the serial dilutions of the K8 scFv was applied and 
the bound antibody fragments were detected with Protein L-HRP. ELISA 
results confirm the dual specific nature of the K8 antibody. 



WO 2004/003019 



PCT/GB2003/002804 



39 



Figure 6 



shows the binding characteristics of the clone OV^/dummy Vh analysed 
using soluble scFv ELISA. Production of the soluble scFv fragments was 
induced by IPTG as described by Harrison et al, Methods Enzymol. 
1996;267:83-109 and the supernatant containing scFv assayed directly. 
Soluble scFv ELISA is performed as described in example 1 and the bound 
scFvs were detected with Protein L-HRP. The ELISA results revealed that 
this clone was still able to bind /3-gal, whereas binding BS A was abolished. 



10 Figure 7 



shows the sequence of variable domain vectors 1 and 2. 



Figure 8 



is a map of the Ch vector used to construct a Vh1/Vh2 multipsecific 
ligand. 



15 Figure 9 



is a map of the V K vector used to construct a V K 1 /V K 2 multispecific ligand. 



Figure 10 TNF receptor assay comparing TAR1-5 dimer 4, TAR1-5-19 dimer 4 and 

TAR1-5-19 monomer. 



20 Figure 1 1 



TNF receptor assay comparing TAR1-5 dimers 1-6. All dimers have been 
FPLC purified and the results for the optimal dimeric species are shown. 



25 



Figure 12 



TNF receptor assay of TAR1-5 19 homodimers in different formats: dAb- 
linker-dAb format with 3U, 5U or 7U linker, Fab format and cysteine 
hinge linker format. 



30 



Figure 13 



4 

Dummy VH sequence for library 1 . The sequence of the VH framework 
based on germline sequence DP47 - JH4b. Positions where NNK 
randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) 
has been incorporated into library 1 are indicated in bold underlined text. 



Figure 14 



Dummy VH sequence for library 2. The sequence of the VH framework 
based on germline sequence DP47 - JH4b. Positions where NNK 



PCT/GB2003/002804 

40 

randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) 
has been incorporated into library 2 are indicated in bold underlined text. 

Dummy Vk sequence for library 3. The sequence of the V/c framework 
based on germline sequence DPk9 - J k1. Positions where NNK 
randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) 
has been incorporated into library 3 are indicated in bold underlined text. 

Nucleotide and amino acid sequence of anti MSA dAbs MSA 16 and MSA 
26. 

Inhibition biacore of MSA 16 and 26. Purified dAbs MSA16 and MSA26 
were analysed by inhibition biacore to determine Kd. Briefly, the dAbs 
were tested to determine the concentration of dAb required to achieve 
200RUs of response on a biacore CM5 chip coated with a high density of 
MSA. Once the required concentrations of dAb had been determined, 
MSA antigen at a range of concentrations around the expected Kd was 
premixed with the dAb and incubated overnight. Binding to the MSA 
coated biacore chip of dAb in each of the premixes was then measured at a 
high flow-rate of 30 /d/minute. 

Serum levels of MSA16 following injection. Serum half life of the dAb 
MSA16 was determined in mouse. MSA16 was dosed as single i.v. 
injections at approx 1.5mg/kg into CD1 mice. Modelling with a 2 
compartment model showed MSA16 had a Mi 2a of 0.98hr, a 1 1/2)8 of 
36.5hr and an AUC of 913hr.mg/ml. MSA16 had a considerably 
lengthened half life compared with HEL4 (an anti-hen egg white lysozyme 
dAb) which had a tl/2aof 0.06hr and atl/2/3 of 0.34hr. 

ELIS A (a) and TNF receptor assay (c) showing inhibition of TNP binding 
with a Fab-like fragment comprising MSA26Ck and TAR1-5-19CH. 
Addition of MSA with the Fab-like fragment reduces the level of 
inhibition. An ELIS A plate coated with l/ig/ml TNFa was probed with 



WO 2004/003019 



PCT/GB2003/002804 



41 

dual specific Vk C h and V/c C/c Fab like fragment and also with a control 
TNFa binding dAb at a concentration calculated to give a similar signal on 
the ELISA. Both the dual specific and control dAb were used to probe the 
ELISA plate in the presence and in the absence of 2mg/ml MSA. The 
signal in the dual specific well was reduced by more than 50% but the 
signal in the dAb well was not reduced at all (see figure 19a). The same 
dual specific protein was also put into the receptor assay with and without 
MSA and competition by MSA was also shown (see figure 19c). This 
demonstrates that binding of MSA to the dual specific is competitive with 
binding to TNFa. 

Figure 20 TNF receptor assay showing inhibiton of TNF binding with a disulphide 

bonded heterodimer of TAR1-5-19 dAb and MSA16 dAb. Addition of 
MSA with the dimer reduces the level of inhibiton in a dose dependant 
manner. The TNF receptor assay (figure 19 (b)) was conducted in the 
presence of a constant concentration of heterodimer (18nM) and a dilution 
series of MSA and HSA. The presence of HSA at a range of 
concentrations (up to 2 mg/ml) did not cause a reduction in the ability of 
the dimer to inhibit TNFa . However, the addition of MSA caused a dose 
dependant reduction in the ability of the dimer to inhibit TNFa (figure 
19a).This demonstrates that MSA and TNFa compete for binding to the 
cys bonded TARl-5^19, MSA16 dimer. MSA and HSA alone did not 
have an effect on the TNF binding level in the assay. 



Detailed Description of the Invention 
Definitions 

Complementary Two immunoglobulin domains are "complementary 55 where they 
belong to families of structures which form cognate pairs or groups or are derived from 
such families and retain this feature. For example, a V H domain and a V L domain of an 
antibody are complementary; two Vh domains are not complementary, and two V L 



WO 2004/003019 PCT/GB2003/002804 

42 

domains are not complementary. Complementary domains may be found in other 
members of the immunoglobulin superfamily, such as the V a and V p (or y and 5) domains 
of the T-cell receptor. In the context of the second configuration of the present invention, 
non-complementary domains do not bind a target molecule cooperatively, but act 
5 independently on different target epitopes which may be on the same or different 
molecules. Domains which are artificial, such as domains based on protein scaffolds 
which do not bind epitopes unless engineered to do so, are non-complementary. 
Likewise, two domains based on (for example) an immunoglobulin domain and a 
fibronectin domain are not complementary. 

10 

Immunoglobulin This refers to a family of polypeptides which retain the 
immunoglobulin fold characteristic of antibody molecules, which contains two 0 sheets 
and, usually, a conserved disulphide bond. Members of the immunoglobulin superfamily 
are involved in many aspects of cellular and non-cellular interactions in vivo, including 
15 widespread roles in the immune system (for example, antibodies, T-cell receptor 
molecules and the like), involvement in cell adhesion (for example the ICAM molecules) 
and intracellular signalling (for example, receptor molecules, such as the PDGF receptor). 
The present invention is applicable to all immunoglobulin superfamily molecules which 
possess binding domains. Preferably, the present invention relates to antibodies. 

20 

Combining Variable domains according to the invention are combined to form a group 
of domains; for example, complementary domains may be combined, such as Vl domains 
being combined with V H domains. Non-complementary domains may also be combined. 
Domains may be combined in a number of ways, involving linkage of the domains by 
25 covalent or non-covalent means. 



Domain A domain is a folded protein structure which retains its tertiary structure 
independently of the rest of the protein. Generally, domains are responsible for discrete 
functional properties of proteins, and in many cases may be added, removed or 
30 transferred to other proteins without loss of function of the remainder of the protein 
and/or of the domain. By single antibody variable domain is meant a folded polypeptide 
domain comprising sequences characteristic of antibody variable domains. It therefore 
includes complete antibody variable domains and modified variable domains, for example 



WO 2004/003019 PCT/GB2003/002804 

43 

in which one or more loops have been replaced by sequences which are not characteristic 
of antibody variable domains, or antibody variable domains which have been truncated or 
comprise N- or C-terminal extensions, as well as folded fragments of variable domains 
which retain at least in part the binding activity and specificity of the full-length domain. 

5 

Repertoire A collection of diverse variants, for example polypeptide variants which 
differ in their primary sequence. A library used in the present invention will encompass a 
repertoire of polypeptides comprising at least 1000 members. 

10 Library The term library refers to a mixture of heterogeneous polypeptides or 
nucleic acids. The library is composed of members, each of which have a single 
polypeptide or nucleic acid sequence. To this extent, library is synonymous with 
repertoire. Sequence differences between library members are responsible for the 
diversity present in the library. The library may take the form of a simple mixture of 

15 polypeptides or nucleic acids, or may be in the form of organisms or cells, for example 
bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic 
acids. Preferably, each individual organism or cell contains only one or a limited number 
of library members. Advantageously, the nucleic acids are incorporated into expression 
vectors, in order to allow expression of the polypeptides encoded by the nucleic acids. In 

20 a preferred aspect, therefore, a library may take the form of a population of host 
organisms, each organism containing one or more copies of an expression vector 
containing a single member of the library in nucleic acid form which can be expressed to 
produce its corresponding polypeptide member. Thus, the population of host organisms 
has the potential to encode a large repertoire of genetically diverse polypeptide variants. 

25 

A c closed conformation multi-specific ligand 5 describes a multi-specific ligand as 
herein defined comprising at least two epitope binding domains as herein defined. The 
term 'closed conformation' (multi-specific ligand) means that the epitope binding 
domains of the ligand are arranged such that epitope binding by one epitope binding 
30 domain competes with epitope binding by another epitope binding domain. That is, 
cognate epitopes may be bound by each epitope binding domain individually but not 
simultaneosuly. The closed conformation of the ligand can be achieved using methods 
herein described. 



WO 2004/003019 



PCT/GB2003/002804 



44 

Antibody An antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such 
as a Fab , F(ab') 2 , Fv, disulphide linked Fv 5 scFv ? closed conformation multispecific 
antibody, disulphide-linked scFv, diabody) whether derived from any species naturally 
producing an antibody, or created by recombinant DNA technology; whether isolated 
5 from serum, B-cells, hybridomas, transfectomas, yeast or bacteria). 

Dual-specific ligand A ligand comprising a first immunoglobulin single variable domain 
and a second immunoglobulin single variable domain as herein defined, wherein the 
variable regions are capable of binding to two different antigens or two epitopes on the 

10 same antigen which are not normally bound by a monospecific immunoglobulin. For 
example, the two epitopes may be on the same hapten, but are not the same epitope or 
sufficiently adjacent to be bound by a monospecific ligand. The dual specific ligands 
according to the invention are composed of variable domains which have different 
specificities, and do not contain mutually complementary variable domain pairs which 

15 have the same specificity. 

Antigen A molecule that is bound by a ligand according to the present invention. 
Typically, antigens are bound by antibody ligands and are capable of raising an antibody 
response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule. 
20 Generally, the dual specific ligands according to the invention are selected for target 
specificity against a particular antigen. In the case of conventional antibodies and 
fragments thereof, the antibody binding site defined by the variable loops (LI, L2, L3 and 
HI, H2, H3) is capable of binding to the antigen. 

25 Epitope A unit of structure conventionally bound by an immunoglobulin Vr/Vl 
pair. Epitopes define the minimum binding site for an antibody, and thus represent the 
target of specificity of an antibody. In the case of a single domain antibody, an epitope 
represents the unit of structure bound by a variable domain in isolation. 

30 Generic ligand A ligand that binds to all members of a repertoire. Generally, not bound 
through the antigen binding site as defined above. Non-limiting examples include protein 
A, protein L and protein G. 



WO 2004/003019 



PCT/GB2003/002804 



45 

Selecting Derived by screening, or derived by a Darwinian selection process, in 
which binding interactions are made between a domain and the antigen or epitope or 
between an antibody and an antigen or epitope. Thus a first variable domain may be 
selected for binding to an antigen or epitope in the presence or in the absence of a 
5 complementary variable domain. 

Universal framework A single antibody framework sequence corresponding to 

the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of 
Proteins of Immunological Interest", US Department of Health and Human Services) or 
10 corresponding to the human germline immunoglobulin repertoire or structure as defined 
by Chothia and Lesk, (1987) J. Mol. Biol. 196:910-917. The invention provides for the 
use of a single framework, or a set of such frameworks, which has been found to permit 
the derivation of virtually any binding specificity though variation in the hypervariable 
regions alone. 

15 

Half-life The time taken for the serum concentration of the ligand to reduce by 50%, 
in vivo, for example due to degradation of the ligand and/or clearance or sequestration of 
the ligand by natural mechanisms. The ligands of the invention are stabilised in vivo and 
their half-life increased by binding to molecules which resist degradation and/or clearance 

20 or sequestration. Typically, such molecules are naturally occurring proteins which 
themselves have a long half-life in vivo. The half-life of a ligand is increased if its 
functional activity persists, in vivo, for a longer period than a similar ligand which is not 
specific for the half-life increasing molecule. Thus, a ligand specific for HSA and a target 
molecule is compared with the same ligand wherein the specificity for HSA is not 

25 present, that it does not bind HSA but binds another molecule. For example, it may bind 
a second epitope on the target molecule. Typically, the half life is increased by 1 0%, 
20%, 30%, 40%, 50% or more. Increases in the range of 2x, 3x, 4x, 5x, lOx, 20x, 30x, 
40x, 5 Ox or more of the half life are possible. Alternatively, or in addition, increases in 
the range of up to 30x, 40x, 50x, 60x, 70x, 80x, 90x, lOOx, 150x of the half life are 

30 possible. 

Homogeneous immunoassay An immunoassay in which analyte is detected 

without need for a step of separating bound and un-bound reagents. 



WO 2004/003019 



PCT/GB2003/002804 



46 

Substantially identical (or "substantially homologous") A first amino acid or 

nucleotide sequence that contains a sufficient number of identical or equivalent (e.g. 9 with 
a similar side chain, e.g., conserved amino acid substitutions) amino acid residues or 
5 nucleotides to a second amino acid or nucleotide sequence such that the first and second 
amino acid or nucleotide sequences have similar activities. In the case of antibodies, the 
second antibody has the same binding specificity and has at least 50% of the affinity of 
the same. 

10 As used herein, the terms "low stringency " "medium stringency/' "high stringency/ 5 
or "very high stringency conditions" describe conditions for nucleic acid hybridization 
and washing. Guidance for performing hybridization reactions can be found in Current 
Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is 
incorporated herein by reference in its entirety. Aqueous and nonaqueous methods are 

15 described in that reference and either can be used. Specific hybridization conditions 
referred to herein are as follows: (1) low stringency hybridization conditions in 6X 
sodium chloride/sodium citrate (SSC) at about 45 °C, followed by two washes in 0.2X 
SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C 
for low stringency conditions); (2) medium stringency hybridization conditions in 6X 

20 SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 60°C; (3) 
high stringency hybridization conditions in 6X SSC at about 45°C, followed by one or 
more washes in 0.2X SSC, 0.1% SDS at 65°C; and preferably (4) very high stringency 
hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one 
or more washes at 0.2X SSC, 1% SDS at 65°C. Very high stringency conditions (4) are 

25 the preferred conditions and the ones that should be used unless otherwise specified. 

Detailed Description of the Invention 

30 Unless defined otherwise, all technical and scientific terms used herein have the same 
meaning as commonly understood by one of ordinary skill in the art (e.g., in cell culture, 
molecular genetics, nucleic acid chemistry, hybridisation techniques and biochemistry). 
Standard techniques are used for molecular, genetic and biochemical methods (see 



WO 2004/003019 



PCT/GB2003/002804 



47 

generally, Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d ed. (1989) 
Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. and Ausubel et al, Short 
Protocols in Molecular Biology (1999) 4 th Ed, John Wiley & Sons, Inc. which are 
incorporated herein by reference) and chemical methods. 



Preparation of immunoglobulin based multi-specific Iigands 

Dual specific Iigands according to the invention, whether open or closed in conformation 
according to the desired configuration of the invention, may be prepared according to 
previously established techniques, used in the field of antibody engineering, for the 
preparation of scFv, "phage" antibodies and other engineered antibody molecules. 
Techniques for the preparation of antibodies, and in particular bispecific antibodies, are 
for example described in the following reviews and the references cited therein: Winter & 
Milstein, (1991) Nature 349:293-299; Plueckthun (1992) Immunological Reviews 
130:151-188; Wright et al, (1992) Crti. Rev. Immunol.l2:125-168; Holliger, P. & 
Winter, G. (1993) Curr. Op. Biotechn. 4, 446-449; Carter, et al. (1995) J. Hematother. 4, 
463-470; Chester, K.A. & Hawkins, RE. (1995) Trends Biotechn. 13, 294-300; 
Hoogenboom, H.R. (1997) Nature Biotechnol. 15, 125-126;' Fearon, D. (1997) Nature 
Biotechnol. 15, 618-619; Pliickthun, A. & Pack, P. (1997) Immunotechnology 3, 83-105; 
Carter, P. & Merchant, A.M. (1997) Curr. Opin. Biotechnol. 8, 449-454; Holliger, P. & 
Winter, G. (1997) Cancer Immunol. Immunother. 45,128-130. 

The invention provides for the selection of variable domains against two different 
antigens or epitopes, and subsequent combination of the variable domains. 

The techniques employed for selection of the variable domains employ libraries and 
selection procedures which are known in the art. Natural libraries (Marks et al (1991) J. 
Mol Biol., 222: 581; Vaughan et al. (1996) Nature Biotech., 14: 309) which use 
rearranged V genes harvested from human B cells are well known to those skilled in the 
art. Synthetic libraries (Hoogenboom & Winter (1992) /. Mol. Biol, 227: 381; Barbas et 
al. (1992) Proc. Natl. Acad. Sci. USA, 89: 4457; Nissim et al. (1994) EMBO J., 13: 692; 
Griffiths et al. (1994) EMBO J., 13: 3245; De Kruif et al. (1995) J. Mol. Biol, 248: 97) 
are prepared by cloning immunoglobulin V genes, usually using PCR. Errors in the PCR 



WO 2004/003019 PCT/GB2003/002804 

48 

process can lead to a high degree of randomisation. Vh and/or Vl libraries may be 
selected against target antigens or epitopes separately, in which case single domain 
binding is directly selected for, or together. 

A preferred method for making a dual specific ligand according to the present invention 
comprises using a selection system in which a repertoire of variable domains is selected 
for binding to a first antigen or epitope and a repertoire of variable domains is selected for 
binding to a second antigen or epitope. The selected variable first and second variable 
domains are then combined and the dual-specific ligand selected for binding to both first 
and second antigen or epitope. Closed conformation ligands are selected for binding both 
first and second antigen or epitope in isolation but not simultaneously. 

A. Library vector systems 

A variety of selection systems are known in the art which are suitable for use in the 
present invention. Examples of such systems are described below. 

Bacteriophage lambda expression systems may be screened directly as bacteriophage 
plaques or as colonies of lysogens, both as previously described (Huse et al (1989,1 
Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl. Acad. Set U.S.A., 87; 
Mullinax et al. (1990) Proc. Natl Acad. Sci. U.S.A., 87: 8095; Persson et al. (1991) Proc. 
Natl. Acad. Sci. U.S.A., 88: 2432) and are of use in the invention. Whilst such expression 
systems can be used to screen up to 10 6 different members of a library, they are not really 
suited to screening of larger numbers (greater than 10 6 members). 

Of particular use in the construction of libraries are selection display systems, which 
enable a nucleic acid to be linked to the polypeptide it expresses. As used herein, a 
selection display system is a system that permits the selection, by suitable display means, 
of the individual members of the library by binding the generic and/or target ligands. 

Selection protocols for isolating desired members of large libraries are known in the art, 
as typified by phage display techniques. Such systems, in which diverse peptide 
sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith 



WO 2004/003019 PCT/GB2003/002804 

49 

(1990) Science, 249: 386), have proven useful for creating libraries of antibody 
fragments (and the nucleotide sequences that encoding them) for the in vitro selection and 
amplification of specific antibody fragments that bind a target antigen (McCafferty et al, 
WO 92/01047). The nucleotide sequences encoding the V H and V L regions are linked to 
5 gene fragments which encode leader signals that direct them to the periplasmic space of 
E. coli and as a result the resultant antibody fragments are displayed on the surface of the 
bacteriophage, typically as fusions to bacteriophage coat proteins (e.g., pill or pVTII). 
Alternatively, antibody fragments are displayed externally on lambda phage capsids 
(phagebodies). An advantage of phage-based display systems is that, because they are 
10 biological systems, selected library members can be amplified simply by growing the 
phage containing the selected library member in bacterial cells. Furthermore, since the 
nucleotide sequence that encode the polypeptide library member is contained on a phage 
or phagemid vector, sequencing, expression and subsequent genetic manipulation is 
relatively straightforward. 

15 

Methods for the construction of bacteriophage antibody display libraries and lambda 
phage expression libraries are well known in the art (McCafferty et al (1990) Nature, 
348: 552; Kang et al (1991) Proc. Natl. Acad. Sci. U.S.A., 88: 4363; Clackson et al 

(1991) Nature, 352: 624; Lowman et al (1991) Biochemistry, 30: 10832; Burton et al 
20 (1991) Proc. Natl Acad. Sci U.S.A., 88: 10134; Hoogenboom et al (1991) Nucleic Acids 

Res., 19: 4133; Chang et al (1991) J. Immunol, 147: 3610; Breitling et al (1991) Gene, 
104: 147; Marks et al (1991) supra; Barbas et al (1992) supra; Hawkins and Winter 

(1992) J. Immunol, 22: 867; Marks et al, 1992, J. Biol Chem., 267: 16007; Lemer et al 
(1992) Science, 258: 1313, incorporated herein by reference). 

25 

One particularly advantageous approach has been the use of scFv phage-libraries (Huston 
et al, 1988, Proc. Natl. Acad. Sci U.S.A., 85: 5879-5883; Chaudhary et al (1990) Proc. 
Natl. Acad. Sci U.S.A., 87: 1066-1070; McCafferty et al (1990) supra; Clackson et al 
(1991) Nature, 352: 624; Marks et al (1991) J. Mol Biol, 222: 581; Chiswell et al 
30 (1992) Trends Biotech., 10: 80; Marks et al (1992) J. Biol Chem., 267). Various 
embodiments of scFv libraries displayed on bacteriophage coat proteins have been 
described. Refinements of phage display approaches are also known, for example as 



WO 2004/003019 



PCT/GB2003/002804 



50 

described in WO96/06213 and WO92/01047 (Medical Research Council et al) and 
WO97/08320 (Morphosys), which are incorporated herein by reference. 

Other systems for generating libraries of polypeptides involve the use of cell-free 
5 enzymatic machinery for the in vitro synthesis of the library members. In one method, 
RNA molecules are selected by alternate rounds of selection against a target ligand and 
PCR amplification (Tuerk and Gold (1990) Science, 249: 505; Ellington and Szostak 
(1990) Nature, 346: 818). A similar technique may be used to identify DNA sequences 
which bind a predetermined human transcription factor (Thiesen and Bach (1990) Nucleic 

10 Acids Res., 18: 3203; Beaudry and Joyce (1992) Science, 257: 635; WO92/05258 and 
W092/14843). In a similar way, in vitro translation can be used to synthesise 
polypeptides as a method for generating large libraries. These methods which generally 
comprise stabilised polysome complexes, are described further in WO88/08453, 
WO90/05785, WO90/07003, WO91/02076, WO91/05058, and WO92/02536. Alternative 

15 display systems which are not phage-based, such as those disclosed in W095/22625 and 
W095/1 1922 (Affymax) use the polysomes to display polypeptides for selection. 

A still further category of techniques involves the selection of repertoires in artificial 
compartments, which allow the linkage of a gene with its gene product. For example, a 

20 selection system in which nucleic acids encoding desirable gene products may be selected 
in microcapsules formed by water-in-oil emulsions is described in WO99/02671, 
WO00/40712 and Tawfik & Griffiths (1998) Nature Biotechnol 16(7), 652-6. Genetic 
elements encoding a gene product having a desired activity are compartmentalised into 
microcapsules and then transcribed and/or translated to produce their respective gene 

25 products (RNA or protein) within the microcapsules. Genetic elements which produce 
gene product having desired activity are subsequently sorted. This approach selects gene 
products of interest by detecting the desired activity by a variety of means. 

30 B. Library Construction . 

Libraries intended for selection, may be constructed using techniques known in the art, 
for example as set forth above, or may be purchased from commercial sources. Libraries 



WO 2004/003019 PCT/GB2003/002804 

51 

which are useful in the present invention are described, for example, in WO99/20749. 
Once a vector system is chosen and one or more nucleic acid sequences encoding 
polypeptides of interest are cloned into the library vector, one may generate diversity 
within the cloned molecules by undertaking mutagenesis prior to expression; 
5 alternatively, the encoded proteins may be expressed and selected, as described above, 
before mutagenesis and additional rounds of selection are performed. Mutagenesis of 
nucleic acid sequences encoding structurally optimised polypeptides is carried out by 
standard molecular methods. Of particular use is the polymerase chain reaction, or PCR, 
(Mullis and Faloona (1987) Methods Enzymol. , 155: 335, herein incorporated by 
10 reference). PCR, which uses multiple cycles of DNA replication catalysed by a 
thermostable, DNA-dependent DNA polymerase to amplify the target sequence of 
interest, is well known in the art. The construction of various antibody libraries has been 
discussed in Winter et ah (1994) Ann. Rev. Immunology 12, 433-55, and references cited 
therein. 

15 

PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at 
least 25 pmol of oligonucleotide primers; it may be advantageous to use a larger amount 
of primer when the primer pool is heavily heterogeneous, as each sequence is represented 
by only a small fraction of the molecules of the pool, and amounts become limiting in the 

20 later amplification cycles. A typical reaction mixture includes: 2jxl of DNA, 25 pmol of 
oligonucleotide primer, 2.5 fxl of 10X PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 
jlxI of 1.25 fiM dNTP, 0.15 \xX (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, 
Foster City, CA) and deionized water to a total volume of 25 jil. Mineral oil is overlaid 
and the PCR is performed using a programmable thermal cycler. The length and 

25 temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in 
accordance to the stringency requirements in effect. Annealing temperature and timing 
are determined both by the efficiency with which a primer is expected to anneal to a 
template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid 
molecules are simultaneously amplified and mutagenised, mismatch is required, at least 

30 in the first round of synthesis. The ability to optimise the stringency of primer annealing 
conditions is well within the knowledge of one of moderate skill in the art. An annealing 
temperature of between 30 °C and 72 °C is used. Initial denaturation of the template 
molecules normally occurs at between 92°C and 99°C for 4 minutes, followed by 20-40 



WO 2004/003019 PCT/GB2003/002804 

52 

cycles consisting of denaturation (94-99°C for 15 seconds to 1 minute), annealing 
(temperature determined as discussed above; 1-2 minutes), and extension (72°C for 1-5 
minutes, depending on the length of the amplified product). Final extension is generally 
for 4 minutes at 72°C, and may be followed by an indefinite (0-24 hour) step at 4°C. 

5 

C. Combining single variable domains 

Domains useful in the invention, once selected, may be combined by a variety of methods 
known in the art, including covalent and non-covalent methods. 

10 

Preferred methods include the use of polypeptide linkers, as described, for example, in 
connection with scFv molecules (Bird et al, (1988) Science 242:423-426). Discussion of 
suitable linkers is provided in Bird et al Science 242, 423-426; Hudson et al , Journal 
Immunol Methods 231 (1999) 177-189; Hudson et al, Proc Nat Acad Sci USA 85, 5879- 
15 5883. Linkers are preferably flexible, allowing the two single domains to interact. One 
linker example is a (Gly 4 Ser) n linker, where n=l to 8, eg, 2, 3, 4, 5 or 7. The linkers used 
in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) PNAS 
(USA) 90:6444-6448). 

20 In one embodiment, the linker employed is not an immunoglobulin hinge region. 

Variable domains may be combined using methods other than linkers. For example, the 
use of disulphide bridges, provided through naturally-occurring or engineered cysteine 
residues, may be exploited to stabilise V h -V h >Vl-Vl or V H -V L dimers (Reiter et al, 
25 (1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable 
domains to improve the "fit" and thus the stability of interaction (Ridgeway et al, (1996) 
Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788). 

Other techniques for joining or stabilising variable domains of immunoglobulins, and in 
30 particular antibody V H domains, may be employed as appropriate. 

In accordance with the present invention, dual specific ligands can be in "closed" 
conformations in solution. A "closed" configuration is that in which the two domains (for 



WO 2004/003019 PCT/GB2003/002804 

53 

example V H and V L ) are present in associated form, such as that of an associated V H -V L 
pair which forms an antibody binding site. For example, scFv may be in a closed 
conformation, depending on the arrangement of the linker used to link the V H and V L 
domains. If this is sufficiently flexible to allow the domains to associate, or rigidly holds 
them in the associated position, it is likely that the domains will adopt a closed 
conformation. 

Similarly, Vh domain pairs and V L domain pairs may exist in a closed conformation. 
Generally, this will be a function of close association of the domains, such as by a rigid 
linker, in the ligand molecule. Ligands in a closed conformation will be unable to bind 
both the molecule which increases the half-life of the ligand and a second target molecule. 
Thus, the ligand will typically only bind the second target molecule on dissociation from 
the molecule which increases the half-life of the ligand. 

Moreover, the construction of V H /V H , Vt/V L or V H /V L dimers without linkers provides 
for competition between the domains. 

Ligands according to the invention may moreover be in an open conformation. In such a 
conformation, the ligands will be able to simultaneously bind both the molecule which 
increases the half-life of the ligand and the second target molecule. Typically, variable 
domains in an open configuration are (in the case of V H -V L pairs) held far enough apart 
for the domains not to interact and form an antibody binding site and not to compete for 
binding to their respective epitopes. In the case of V h /Vh or ViTVl dimers, the domains 
are not forced together by rigid linkers. Naturally, such domain pairings will not compete 
for antigen binding or form an antibody binding site. 

Fab fragments and whole antibodies will exist primarily in the closed conformation, 
although it will be appreciated that open and closed dual specific ligands are likely to 
exist in a variety of equilibria under different circumstances. Binding of the ligand to a 
target is likely to shift the balance of the equilibrium towards the open configuration. 
Thus, certain ligands according to the invention can exist in two conformations in 
solution, one of which (the open form) can bind two antigens or epitopes independently, 



WO 2004/003019 PCT/GB2003/002804 

54 

whilst the alternative conformation (the closed form) can only bind one antigen or 
epitope; antigens or epitopes thus compete for binding to the ligand in this conformation. 

Although the open form of the dual specific ligand may thus exist in equilibrium with the 
5 closed form in solution, it is envisaged that the equilibrium will favour the closed form; 
moreover, the open form can be sequestered by target binding into a closed conformation. 
Preferably, therefore, certain dual specific ligands of the invention are present in an 
equilibrium between two (open and closed) conformations. 

10 Dual specific ligands according to the invention may be modified in order to favour an 
open or closed conformation. For example, stabilisation of V h ~Vl interactions with 
disulphide bonds stabilises the closed conformation. Moreover, linkers used to join the 
domains, including Vh domain and Vl domain pairs, may be constructed such that the 
open from is favoured; for example, the linkers may sterically hinder the association of 

15 the domains, such as by incorporation of large amino acid residues in opportune 
locations, or the designing of a suitable rigid structure which will keep the domains 
physically spaced apart. 

D. Characterisation of the dual-specific ligand . 

20 

The binding of the dual-specific ligand to its specific antigens or epitopes can be tested by 
methods which will be familiar to those skilled in the art and include ELISA. In a 
preferred embodiment of the invention binding is tested using monoclonal phage ELISA. 

25 Phage ELISA may be performed according to any suitable procedure: an exemplary 
protocol is set forth below. 

Populations of phage produced at each round of selection can be screened for binding by 
ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. 
30 Phage from single infected bacterial colonies from these populations can then be screened 
by ELISA to identify "monoclonal" phage antibodies. It is also desirable to screen soluble 
antibody fragments for binding to antigen or epitope, and this can also be undertaken by 



WO 2004/003019 PCT/GB2003/002804 

55 

ELISA using reagents, for example, against a C- or N-terminal tag (see for example 
Winter et al (1994) Aim. Rev. Immunology 12, 433-55 and references cited therein. 

The diversity of the selected phage monoclonal antibodies may also be assessed by gel 
electrophoresis of PCR products (Marks et al 1991, supra; Nissim et al. 1994 supra), 
probing (Tomlinson et al, 1992) J. Mol. Biol. 227, 776) or by sequencing of the vector 
DNA. 

E. Structure of 'Dual-specific ligands' . 

As described above, an antibody is herein defined as an antibody (for example IgG, IgM, 
IgA, IgA, IgE) or fragment (Fab, Fv, disulphide linked Fv, scFv, diabody) which 
comprises at least one heavy and a light chain variable domain, at least two heavy chain 
variable domains or at least two light chain variable domains. It may be at least partly 
derived from any species naturally producing an antibody, or created by recombinant 
DNA technology; whether isolated from serum, B~cells, hybridomas, transfectomas, yeast 
or bacteria). 

In a preferred embodiment of the invention the dual-specific ligand comprises at least one 
single heavy chain variable domain of an antibody and one single light chain variable 
domain of an antibody, or two single heavy or light chain variable domains. For example, 
the ligand may comprise a V H /V L pair, a pair of V H domains or a pair of V L domains. 

The first and the second variable domains of such a ligand may be on the same 
polypeptide chain. Alternatively they may be on separate polypeptide chains. In the case 
that they are on the same polypeptide chain they may be linked by a linker, which is 
preferentially a peptide sequence, as described above. 

The first and second variable domains may be covalently or non-covalently associated. In 
the case that they are covalently associated, the covalent bonds may be disulphide bonds. 

In the case that the variable domains are selected from V-gene repertoires selected for 
instance using phage display technology as herein described, then these variable domains 



WO 2004/003019 PCT/GB2003/002804 

56 

comprise a universal framework region, such that is they may be recognised by a specific 
generic ligand as herein defined. The use of universal frameworks, generic ligands and 
the like is described in WO99/20749. 

Where V-gene repertoires are used variation in polypeptide sequence is preferably located 
within the structural loops of the variable domains. The polypeptide sequences of either 
variable domain may be altered by DNA shuffling or by mutation in order to enhance the 
interaction of each variable domain with its complementary pair. DNA shuffling is 
known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and 
U.S. Patent No. 6,297,053, both of which are incorporated herein by reference. Other 
methods of mutagenesis are well known to those of skill in the art. 

In a preferred embodiment of the invention the 'dual-specific ligand' is a single chain Fv 
fragment. In an alternative embodiment of the invention, the 'dual-specific ligand 5 
consists of a Fab format. 

In a further aspect, the present invention provides nucleic acid encoding at least a 'dual- 
specific ligand' as herein defined. 

One skilled in the art will appreciate that, depending on the aspect of the invention, both 
antigens or epitopes may bind simultaneously to the same antibody molecule. 
Alternatively, they may compete for binding to the same antibody molecule. For 
example, where both epitopes are bound simultaneously, both variable domains of a dual 
specific ligand are able to independently bind their target epitopes. Where the domains 
compete, the one variable domain is capable of binding its target, but not at the same time 
as the other variable domain binds its cognate target; or the first variable domain is 
capable of binding its target, but not at the same time as the second variable domain binds 
its cognate target. 

The variable regions may be derived from antibodies directed against target antigens or 
epitopes. Alternatively they may be derived from a repertoire of single antibody domains 
such as those expressed on the surface of filamentous bacteriophage. Selection may be 
performed as described below. 



WO 2004/003019 



57 



PCT/GB2003/002804 



In general, the nucleic acid molecules and vector constructs required for the performance 
of the present invention may be constructed and manipulated as set forth in standard 
laboratory manuals, such as Sambrook et aL (1989) Molecular Cloning: A Laboratory 
5 Manual, Cold Spring Harbor, USA. 

The manipulation of nucleic acids useful in the present invention is typically carried out 
in recombinant vectors. 

10 Thus in a further aspect, the present invention provides a vector comprising nucleic acid 
encoding at least a ' dual-specific ligand 5 as herein defined. 

As used herein, vector refers to a discrete element that is used to introduce heterologous 
DNA into cells for the expression and/or replication thereof. Methods by which to select 

15 or construct and, subsequently, use such vectors are well known to one of ordinary skill in 
the art. Numerous vectors are publicly available, including bacterial plasmids, 
bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used 
for simple cloning and mutagenesis; alternatively gene expression vector is employed. A 
vector of use according to the invention may be selected to accommodate a polypeptide 

20 coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb or more in 
length A suitable host cell is transformed with the vector after in vitro cloning 
manipulations. Each vector contains various functional components, which generally 
include a cloning (or "polylinker") site, an origin of replication and at least one selectable 
marker gene. If given vector is an expression vector, it additionally possesses one or more 

25 of the following: enhancer element, promoter, transcription termination and signal 
sequences, each positioned in the vicinity of the cloning site, such that they are 
operatively linked to the gene encoding a ligand according to the invention. 

Both cloning and expression vectors generally contain nucleic acid sequences that enable 
30 the vector to replicate in one or more selected host cells. Typically in cloning vectors, this 
sequence is one that enables the vector to replicate independently of the host 
chromosomal DNA and includes origins of replication or autonomously replicating 
sequences. Such sequences are well known for a variety of bacteria, yeast and viruses. 



WO 2004/003019 PCT/GB2003/002804 

58 

The origin of replication from the plasmid pBR322 is suitable for most Gram-negative 
bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g. 
SV 40, adenovirus) are useful for cloning vectors in mammalian cells. Generally, the 
origin of replication is not needed for mammalian expression vectors unless these are 
5 used in mammalian cells able to replicate high levels of DNA, such as COS cells. 

Advantageously, a cloning or expression vector may contain a selection gene also 
referred to as selectable marker. This gene encodes a protein necessary for the survival or 
growth of transformed host cells grown in a selective culture medium. Host cells not 
10 transformed with the vector containing the selection gene will therefore not survive in the 
culture medium. Typical selection genes encode proteins that confer resistance to 
antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, 
complement auxotrophic deficiencies, or supply critical nutrients not available in the 
growth media. 

15 

Since the replication of vectors encoding a ligand according to the present invention is 
most conveniently performed in E. coli, an E, co/z-selectable marker, for example, the p~ 
lactamase gene that confers resistance to the antibiotic ampicillin, is of use. These can be 
obtained from E. coli plasmids, such as pBR322 or a pUC plasmid such as pUC18 or 
20 pUC19. 

Expression vectors usually contain a promoter that is recognised by the host organism and 
is operably linked to the coding sequence of interest. Such a promoter may be inducible 
or constitutive. The term "operably linked" refers to a juxtaposition wherein the 
25 components described are in a relationship permitting them to function in their intended 
manner. A control sequence "operably linked" to a coding sequence is ligated in such a 
way that expression of the coding sequence is achieved under conditions compatible with 
the control sequences. 

30 Promoters suitable for use with prokaryotic hosts include, for example, the p-lactamase 
and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system 
and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems 



WO 2004/003019 PCT/GB2003/002804 

59 

will also generally contain a Shine-Delgarno sequence operably linked to the coding 
sequence. 

The preferred vectors are expression vectors that enables the expression of a nucleotide 
sequence corresponding to a polypeptide library member. Thus, selection with the first 
and/or second antigen or epitope can be performed by separate propagation and 
expression of a single clone expressing the polypeptide library member or by use of any 
selection display system. As described above, the preferred selection display system is 
bacteriophage display. Thus, phage or phagemid vectors may be used, eg pITl or pIT2. 
Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pelA. 
One example are phagemid vectors which have an E. colt origin of replication (for 
double stranded replication) and also a phage origin of replication (for production of 
single-stranded DNA). The manipulation and expression of such vectors is well known in 
the art (Hoogenboom and Winter (1992) supra; Nissim et ah (1994) supra). Briefly, the 
vector contains a (3-lactamase gene to confer selectivity on the phagemid and a lac 
promoter upstream of a expression cassette that consists (N to C terminal) of a pelB 
leader sequence (which directs the expressed polypeptide to the periplasmic space), a 
multiple cloning site (for cloning the nucleotide version of the library member), 
optionally, one or more peptide tag (for detection), optionally, one or more TAG stop 
codon and the phage protein pill. Thus, using various suppressor and non-suppressor 
strains of E. coli and with the addition of glucose, iso-propyl thio-p-D-galactoside (EPTG) 
or a helper phage, such as VCS Ml 3, the vector is able to replicate as a plasmid with no 
expression, produce large quantities of the polypeptide library member only or produce 
phage, some of which contain at least one copy of the polyp eptide-pIII fusion on their 
surface. 

Construction of vectors encoding ligands according to the invention employs 
conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, 
tailored, and religated in the form desired to generate the required vector. If desired, 
analysis to confirm that the correct sequences are present in the constructed vector can be 
performed in a known fashion. Suitable methods for constructing expression vectors, 
preparing in vitro transcripts, introducing DNA into host cells, and performing analyses 
for assessing expression and function are known to those skilled in the art. The presence 



WO 2004/003019 PCT/GB2003/002804 

60 

of a gene sequence in a sample is detected, or its amplification and/or expression 
quantified by conventional methods, such as Southern or Northern analysis, Western 
blotting, dot blotting of DNA, RNA or protein, in situ hybridisation, 
immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those 
5 skilled in the art will readily envisage how these methods may be modified, if desired. 

Structure of closed conformation multisyecific ligands 

According to one aspect of the second configuration of the invention present invention, 
10 the two or more non-complementary epitope binding domains are linked so that they are 
in a closed conformation as herein defined. Advantageously, they may be further 
attached to a skeleton which may, as a alternative, or on addition to a linker described 
herein, facilitate the formation and/or maintenance of the closed conformation of the 
epitope binding sites with respect to one another. 

15 

(I) Skeletons 

Skeletons may be based on immunoglobulin molecules or may be non-immunoglobulin in 
origin as set forth above. Preferred immunoglobulin skeletons as herein defined includes 
any one or more of those selected from the following: an immunoglobulin molecule 

20 comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the 
CHI domain of an antibody heavy chain; an immunoglobulin molecule comprising the 
CHI and CH2 domains of an antibody heavy chain; an immunoglobulin molecule 
comprising the CHI, CH2 and CH3 domains of an antibody heavy chain; or any of the 
subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody. 

25 A hinge region domain may also be included.. Such combinations of domains may, for 
example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, 
scFv, Fab or F(ab')2 molecules. Those skilled in the art will be aware that this list is not 
intended to be exhaustive. 

30 (II) Protein scaffolds 

Each epitope binding domain comprises a protein scaffold and one or more CDRs which 
are involved in the specific interaction of the domain with one or more epitopes. 
Advantageously, an epitope binding domain according to the present invention comprises 



WO 2004/003019 PCT/GB2003/002804 

61 

three CDRs. Suitable protein scaffolds include any of those selected from the group 
consisting of the following: those based on immunoglobulin domains, those based on 
fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones 
such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors 
5 SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be 
exhaustive. 

F: Scaffolds for use in Constructing Dual Specific Ligands 

10 i. Selection of the main-chain conformation 

The members of the immunoglobulin superfamily all share a similar fold for their 
polypeptide chain. For example, although antibodies are highly diverse in terms of their 
primary sequence, comparison of sequences and crystallo graphic structures has revealed 
that, contrary to expectation, five of the six antigen binding loops of antibodies (HI, H2, 

15 LI, L2, L3) adopt a limited number of main-chain conformations, or canonical structures 
(Chothia and Lesk (1987) J. Mol Biol, 196: 901; Chothia et al (1989) Nature, 342: 877). 
Analysis of loop lengths and key residues has therefore enabled prediction of the main- 
chain conformations of HI, H2, LI, L2 and L3 found in the majority of human antibodies 
(Chothia et al (1992) J. Mol Biol, 227: 799; Tomlinson et al (1995) EMBO J., 14: 

20 4628; Williams et al (1996) J. Mol Biol, 264: 220). Although the H3 region is much 
more diverse in terms of sequence, length and structure (due to the use of D segments), it 
also forms a limited number of main-chain conformations for short loop lengths which 
depend on the length and the presence of particular residues, or types of residue, at key 
positions in the loop and the antibody framework (Martin et al (1996) J. Mol Biol, 263: 

25 800; Shirai et al (1996) FEBS Letters, 399: 1). 

The dual specific ligands of the present invention are advantageously assembled from 
libraries of domains, such as libraries of Vh domains and/or libraries of V L domains. 
Moreover, the dual specific ligands of the invention may themselves be provided in the 
30 form of libraries. In one aspect of the present invention, libraries of dual specific ligands 
and/or domains are designed in which certain loop lengths and key residues have been 
chosen to ensure that the main-chain conformation of the members is known. 
Advantageously, these are real conformations of immunoglobulin superfamily molecules 



WO 2004/003019 PCT/GB2003/002804 

62 

found in nature, to minimise the chances that they are non-functional, as discussed 
above. Germline V gene segments serve as one suitable basic framework for constructing 
antibody or T-cell receptor libraries; other sequences are also of use. Variations may 
occur at a low frequency, such that a small number of functional members may possess an 
altered main-chain conformation, which does not affect its function. 

Canonical structure theory is also of use to assess the number of different main-chain 
conformations encoded by ligands, to predict the main-chain conformation based on 
ligand sequences and to chose residues for diversification which do not affect the 
canonical structure. It is known that, in the human V K domain, the LI loop can adopt one 
of four canonical structures, the L2 loop has a single canonical structure and that 90% of 
human V K domains adopt one of four or five canonical structures for the L3 loop 
(Tomlinson et al (1995) supra); thus, in the V K domain alone, different canonical 
structures can combine to create a range of different main-chain conformations. Given 
that the V x domain encodes a different range of canonical structures for the LI, L2 and L3 
loops and that V K and V* domains can pair with any V H domain which can encode several 
canonical structures for the HI and H2 loops, the number of canonical structure 
combinations observed for these five loops is very large. This implies that the generation 
of diversity in the main-chain conformation may be essential for the production of a wide 
range of binding specificities. However, by constructing an antibody library based on a 
single known main-chain conformation it has been found, contrary to expectation, that 
diversity in the main-chain conformation is not required to generate sufficient diversity to 
target substantially all antigens. Even more surprisingly, the single main-chain 
conformation need not be a consensus structure - a single naturally occurring 
conformation can be used as the basis for an entire library. Thus, in a preferred aspect, the 
dual-specific ligands of the invention possess a single known main-chain conformation. 

The single main-chain conformation that is chosen is preferably commonplace among 
molecules of the immunoglobulin superfamily type in question. A conformation is 
commonplace when a significant number of naturally occurring molecules are observed 
to adopt it. Accordingly, in a preferred aspect of the invention, the natural occurrence of 
the different main-chain conformations for each binding loop of an immunoglobulin 
domain are considered separately and then a naturally occurring variable domain is 



WO 2004/003019 PCT/GB2003/002804 

63 

chosen which possesses the desired combination of main-chain conformations for the 
different loops. If none is available, the nearest equivalent may be chosen. It is preferable 
that the desired combination of main-chain conformations for the different loops is 
created by selecting germline gene segments which encode the desired main-chain 
5 conformations. It is more preferable, that the selected germline gene segments are 
frequently expressed in nature, and most preferable that they are the most frequently 
expressed of all natural germline gene segments. 



In designing dual specific ligands or libraries thereof the incidence of the different main- 

10 chain conformations for each of the six antigen binding loops may be considered 
separately. For HI, H2, LI, L2 and L3, a given conformation that is adopted by between 
20% and 100% of the antigen binding loops of naturally occurring molecules is chosen. 
Typically, its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, 
above 50% or even above 65%. Since the vast majority of H3 loops do not have canonical 

15 structures, it is preferable to select a main-chain conformation which is commonplace 
among those loops which do display canonical structures. For each of the loops, the 
conformation which is observed most often in the natural repertoire is therefore selected. 
In human antibodies, the most popular canonical structures (CS) for each loop are as 
follows: HI - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), LI - CS 2 of 

20 V K (39%), L2 - CS 1 (100%), L3 - CS 1 of V K (36%) (calculation assumes a k:A, ratio of 
70:30, Hood et al (1967) Cold Spring Harbor Symp. Quant. BioL, 48: 133). For H3 loops 
that have canonical structures, a CDR3 length (Kabat et al. (1991) Sequences of proteins 
of immunological interest, U.S. Department of Health and Human Services) of seven 
residues with a salt-bridge from residue 94 to residue 101 appears to be the most 

25 common. There are at least 16 human antibody sequences in the EMBL data library with 
the required H3 length and key residues to form this conformation and at least two 
crystallographic structures in the protein data bank which can be used as a basis for 
antibody modelling (2cgr and ltet). The most frequently expressed germline gene 
segments that this combination of canonical structures are the Vh segment 3-23 (DP-47), 

30 the J H segment JH4b, the V K segment 02/012 (DPK9) and the J K segment J K 1. V H 
segments DP45 and DP38 are also suitable. These segments can therefore be used in 
combination as a basis to construct a library with the desired single main-chain 
conformation. 



WO 2004/003019 



64 



PCT/GB2003/002804 



Alternatively, instead of choosing the single main-chain conformation based on the 
natural occurrence of the different main-chain conformations for each of the binding 
loops in isolation, the natural occurrence of combinations of main-chain conformations is 
5 used as the basis for choosing the single main-chain conformation. In the case of 
antibodies, for example, the natural occurrence of canonical structure combinations for 
any two, three, four, five or for all six of the antigen binding loops can be determined. 
Here, it is preferable that the chosen conformation is commonplace in naturally occurring 
antibodies and most preferable that it observed most frequently in the natural repertoire. 
10 Thus, in human antibodies, for example, when natural combinations of the five antigen 
binding loops, HI, H2, LI, L2 and L3, are considered, the most frequent combination of 
canonical structures is determined and then combined with the most popular 
conformation for the H3 loop, as a basis for choosing the single main-chain conformation. 

15 ii. Diversification of the canonical sequence 

Having selected several known main-chain conformations or, preferably a single 
known main-chain conformation, dual specific ligands according to the invention or 
libraries for use in the invention can be constructed by varying the binding site of the 
molecule in order to generate a repertoire with structural and/or functional diversity. This 

20 means that variants are generated such that they possess sufficient diversity in their 
structure and/or in their function so that they are capable of providing a range of 
activities. 

The desired diversity is typically generated by varying the selected molecule at one or 
25 more positions. The positions to be changed can be chosen at random or are preferably 
selected. The variation can then be achieved either by randomisation, during which the 
resident amino acid is replaced by any amino acid or analogue thereof, natural or 
synthetic, producing a very large number of variants or by replacing the resident amino 
acid with one or more of a defined subset of amino acids, producing a more limited 
30 number of variants. 

Various methods have been reported for introducing such diversity. Error-prone PGR 
(Hawkins et al (1992) J. Mol Biol., 226: 889), chemical mutagenesis (Deng et al (1994) 



WO 2004/003019 PCT/GB2003/002804 

65 

J. Biol Chem., 269: 9533) or bacterial mutator strains (Low et al (1996) J. Mol Biol, 
260: 359) can be used to introduce random mutations into the genes that encode the 
molecule. Methods for mutating selected positions are also well known in the art and 
include the use of mismatched oligonucleotides or degenerate oligonucleotides, with or 
5 without the use of PGR. For example, several synthetic antibody libraries have been 
created by targeting mutations to the antigen binding loops. The H3 region of a human 
tetanus toxoid-binding Fab has been randomised to create a range of new binding 
specificities (Barbas et al. (1992) Proc. Natl Acad. Set USA, 89: 4457). Random or 
semi-random H3 and L3 regions have been appended to germline V gene segments to 

10 produce large libraries with unmutated framework regions (Hoogenboom & Winter 
(1992) J. Mol Biol, 227: 381; Barbas et al. (1992) Proc. Natl Acad. Sci. USA, 89: 4457; 
Nissim et al (1994) EMBO J., 13: 692; Griffiths et al. (1994) EMBO J., 13: 3245; De 
Kruif et al (1995) J. Mol Biol, 248: 97). Such diversification has been extended to 
include some or all of the other antigen binding loops (Crameri et al. (1996) Nature Med., 

15 2: 100; Riechmann et al (1995) Bio/Technology, 13: 475; Morphosys, WO97/08320, 
supra). 

Since loop randomisation has the potential to create approximately more than 10 15 
structures for H3 alone and a similarly large number of variants for the other five loops, it 
20 is not feasible using current transformation technology or even by using cell free systems 
to produce a library representing all possible combinations. For example, in one of the 
largest libraries constructed to date, 6 x 10 10 different antibodies, which is only a fraction 
of the potential diversity for a library of this design, were generated (Griffiths et al. 
(1994) supra). 

25 

In a preferred embodiment, only those residues which are directly involved in creating or 
modifying the desired function of the molecule are diversified. For many molecules, the 
function will be to bind a target and therefore diversity should be concentrated in the 
target binding site, while avoiding changing residues which are crucial to the overall 
30 packing of the molecule or to maintaining the chosen main-chain conformation. 



WO 2004/003019 PCT/GB2003/002804 

66 

Diversification of the canonical sequence as it applies to antibody domains 

In the case of antibody dual-specific ligands, the binding site for the target is most 
often the antigen binding site. Thus, in a highly preferred aspect, the invention provides 
libraries of or for the assembly of antibody dual-specific ligands in which only those 
5 residues in the antigen binding site are varied. These residues are extremely diverse in the 
human antibody repertoire and are known to make contacts in high-resolution 
antibody/antigen complexes. For example, in L2 it is known that positions 50 and 53 are 
diverse in naturally occurring antibodies and are observed to make contact with the 
antigen. In contrast, the conventional approach would have been to diversify all the 
10 residues in the corresponding Complementarity Determining Region (CDR1) as defined 
by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the 
library for use according to the invention. This represents a significant improvement in 
terms of the functional diversity required to create a range of antigen binding specificities. 

15 In nature, antibody diversity is the result of two processes: somatic recombination of 
germline V, D and J gene segments to create a naive primary repertoire (so called 
germline and junctional diversity) and somatic hypermutation of the resulting rearranged 
V genes. Analysis of human antibody sequences has shown that diversity in the primary 
repertoire is focused at the centre of the antigen binding site whereas somatic 

20 hypermutation spreads diversity to regions at the periphery of the antigen binding site that 
are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. Mol. Biol, 
256: 813). This complementarity has probably evolved as an efficient strategy for 
searching sequence space and, although apparently unique to antibodies, it can easily be 
applied to other polypeptide repertoires. The residues which are varied are a subset of 

25 those that form the binding site for the target. Different (including overlapping) subsets of 
residues in the target binding site are diversified at different stages during selection, if 
desired. 

In the case of an antibody repertoire, an initial 'naive 5 repertoire is created where some, 
30 but not all, of the residues in the antigen binding site are diversified. As used herein in 
this context, the term "naive" refers to antibody molecules that have no pre-determined 
target. These molecules resemble those which are encoded by the immunoglobulin genes 
of an individual who has not undergone immune diversification, as is the case with fetal 



WO 2004/003019 PCT/GB2003/002804 

67 

and newborn individuals, whose immune systems have not yet been challenged by a 
wide variety of antigenic stimuli. This repertoire is then selected against a range of 
antigens or epitopes. If required, further diversity can then be introduced outside the 
region diversified in the initial repertoire. This matured repertoire can be selected for 
5 modified function, specificity or affinity. 



The invention provides two different naive repertoires of binding domains for the 
construction of dual specific ligands, or a naive library of dual specific ligands, in which 
some or all of the residues in the antigen binding site are varied. The "primary" library 

10 mimics the natural primary repertoire, with diversity restricted to residues at the centre of 
the antigen binding site that are diverse in the germline V gene segments (germline 
diversity) or diversified during the recombination process (junctional diversity). Those 
residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, 
H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96. In the "somatic" 

15 library, diversity is restricted to residues that are diversified during the recombination 
process (junctional diversity) or are highly somatically mutated). Those residues which 
are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, 
L31, L32, L34 and L96. All the residues listed above as suitable for diversification in 
these libraries are known to make contacts in one or more antibody-antigen complexes. 

20 Since in both libraries, not all of the residues in the antigen binding site are varied, 
additional diversity is incorporated during selection by varying the remaining residues, if 
it is desired to do so. It shall be apparent to one skilled in the art that any subset of any of 
these residues (or additional residues which comprise the antigen binding site) can be 
used for the initial and/or subsequent diversification of the antigen binding site. 

25 

In the construction of libraries for use in the invention, diversification of chosen positions 
is typically achieved at the nucleic acid level, by altering the coding sequence which 
specifies the sequence of the polypeptide such that a number of possible amino acids (all 
20 or a subset thereof) can be incorporated at that position. Using the IUPAC 
30 nomenclature, the most versatile codon is NNK, which encodes all amino acids as well as 
the TAG stop codon. The NNK codon is preferably used in order to introduce the 
required diversity. Other codons which achieve the same ends are also of use, including 



WO 2004/003019 PCT/GB2003/002804 

68 

the NNN codon, which leads to the production of the additional stop codons TGA and 
TAA. 

A feature of side-chain diversity in the antigen binding site of human antibodies is a 
5 pronounced bias which favours certain amino acid residues. If the amino acid 
composition of the ten most diverse positions in each of the Vh, V k and Vx regions are 
summed, more than 76% of the side-chain diversity comes from only seven different 
residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), 
alanine (7%), aspartate (6%) and threonine (6%). This bias towards hydrophilic residues 
10 and small residues which can provide main-chain flexibility probably reflects the 
evolution of surfaces which are predisposed to binding a wide range of antigens or 
epitopes and may help to explain the required promiscuity of antibodies in the primary 
repertoire. 

15 Since it is preferable to mimic this distribution of amino acids, the distribution of amino 
acids at the positions to be varied preferably mimics that seen in the antigen binding site 
of antibodies. Such bias in the substitution of amino acids that permits selection of certain 
polypeptides (not just antibody polypeptides) against a range of target antigens is easily 
applied to any polypeptide repertoire. There are various methods for biasing the amino 

20 acid distribution at the position to be varied (including the use of tri-nucleotide 
mutagenesis, see WO97/08320), of which the preferred method, due to ease of synthesis, 
is the use of conventional degenerate codons. By comparing the amino acid profile 
encoded by all combinations of degenerate codons (with single, double, triple and 
quadruple degeneracy in equal ratios at each position) with the natural amino acid use it is 

25 possible to calculate the most representative codon. The codons (AGT)(AGC)T, 
(AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using 
IUPAC nomenclature - are those closest to the desired amino acid profile: they encode 
22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and 
cysteine. Preferably, therefore, libraries are constructed using either the DVT, DVC or 

30 DVY codon at each of the diversified positions. 



WO 2004/003019 

69 

G: Antigens capable of increasing ligand half-life 



PCT/GB2003/002804 



The dual specific ligands according to the invention, in one configuration thereof, are 
capable of binding to one or more molecules which can increase the half-life of the ligand 
5 in vivo. Typically, such molecules are polypeptides which occur naturally in vivo and 
which resist degradation or removal by endogenous mechanisms which remove unwanted 
material from the organism. For example, the molecule which increases the half-life of 
the organism may be selected from the following: 

10 Proteins from the extracellular matrix; for example collagen, laminins, integrins and 

fibronectin. Collagens are the major proteins of the extracellular matrix. About 15 types 
of collagen molecules are currently known, found in different parts of the body, eg type I 
collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, 
cornea, internal organs or type II collagen found in cartilage, invertebral disc, notochord, 

1 5 vitreous humour of the eye. 

Proteins found in blood, including: 

Plasma proteins such as fibrin, a-2 macroglobulin, serum albumin, fibrinogen A, 
20 fibrinogen B, serum amyloid protein A, heptaglobin, profilin, ubiquitin, uteroglobulin and 
/3-2-micro globulin; 

Enzymes and inhibitors such as plasminogen, lysozyme, cystatin C, alpha- 1 -antitrypsin 
and pancreatic trypsin inhibitor. Plasminogen is the inactive precursor of the trypsin-like 
25 serine protease plasmin. It is normally found circulating through the blood stream. When 
plasminogen becomes activated and is converted to plasmin, it unfolds a potent enzymatic 
domain that dissolves the fibrinogen fibers that entgangle the blood cells in a blood clot. 
This is called fibrinolysis. 

30 Immune system proteins, such as IgE, IgG, IgM. 



Transport proteins such as retinol binding protein, ol-1 microglobulin. 



WO 2004/003019 PCT/GB2003/002804 

70 

Defensins such as beta-defensin 1 , Neutrophil defensins 1 ,2 and 3 . 



Proteins found at the blood brain barrier or in neural tissues, such as melanocortin 
receptor, myelin, ascorbate transporter. 

5 

Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see 
US5977307); 

brain capillary endothelial cell receptor, transferrin, transferrin receptor, insulin, insulin- 
10 like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, 
insulin receptor. 

Proteins localised to the kidney, such as polycystin, type IV collagen, organic anion 
transporter Kl, Heymann's antigen. 

15 

Proteins localised to the liver, for example alcohol dehydrogenase, G250. 

Blood coagulation factor X 
al antitrypsin 
20 HNF la 

Proteins localised to the lung, such as secretory component (binds IgA). 

Proteins localised to the Heart, for example HSP 27. This is associated with dilated 

* 

25 cardiomyopathy. 

Proteins localised to the skin, for example keratin. 

Bone specific proteins, such as bone morphogenic proteins (BMPs), which are a subset of 
30 the transforming growth factor (3 superfamily that demonstrate osteogenic activity. 

Examples include BMP-2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-1) and 
-8 (OP-2). 



WO 2004/003019 PCT/GB2003/002804 

71 

Tumour specific proteins, including human trophoblast antigen, herceptin receptor, 
oestrogen receptor, cathepsins eg cathepsin B (found in liver and spleen). 

Disease-specific proteins, such as antigens expressed only on activated T-cells: including 
LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL) see Nature 402, 
304-309; 1999, OX40 (a member of the TNF receptor family, expressed on activated T 
cells and the only costimulatory T cell molecule known to be specifically up-regulated in 
human T cell leukaemia virus type-I (HTLV-I)-producing cells.) See J Immunol 2000 
Jul l;165(l):263-70\ Metalloproteases (associated with arthritis/cancers), including 
CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; 
angiogenic growth factors, including acidic fibroblast growth factor (FGF-1), basic 
fibroblast growth factor (FGF-2), Vascular endothelial growth factor / vascular 
permeability factor (VEGF/VPF), transforming growth factor-a (TGF a), tumor necrosis 
factor-alpha (TNF-o:), angiogenin, interleukin-3 (IL-3), interleukin-8 (IL-8), platelet- 
derived endothelial growth factor (PD-ECGF), placental growth factor (P1GF), midkine 
platelet-derived growth factor-BB (PDGF), fractalkine. 

Stress proteins (heat shock proteins) 

HSPs are normally found intracellularly. When they are found extracellularly, it is an 
indicator that a cell has died and spilled out its contents. This unprogrammed cell death 
(necrosis) only occurs when as a result of trauma, disease or injury and therefore in vivo, 
extracellular HSPs trigger a response from the immune system that will fight infection 
and disease. A dual specific which binds to extracellular HSP can be localised to a 
disease site. 

Proteins involved in Fc transport 
Brambell receptor (also known as FcRB) 

This Fc receptor has two functions, both of which are potentially useful for delivery 
The functions are 

(1) The transport of IgG from mother to child across the placenta 

(2) the protection of IgG from degradation thereby prolonging its serum half life of 
IgG. It is thought that the receptor recycles IgG from endosome. 



WO 2004/003019 

72 

See Holliger et al 9 Nat Biotechnol 1997 Jul;15(7):632-6 



PCT/GB2003/002804 



Ligands according to the invention may designed to be specific for the above targets 
without requiring any increase in or increasing half life in vivo. For example, ligands 
5 according to the invention can be specific for targets selected from the foregoing which 
are tissue-specific, thereby enabling tissue-specific targeting of the dual specific ligand, 
or a dAb monomer that binds a tissue-specific therapeutically relevant target, irrespective 
of any increase in half-life, although this may result. Moreover, where the ligand or dAb 
monomer targets kidney or liver, this may redirect the ligand or dAb monomer to an 
10 alternative clearance pathway in vivo (for example, the ligand may be directed away from 
liver clearance to kidney clearance). 

H: Use of multispecific ligands according to the second configuration of the 
invention 

15 

Multispecific ligands according to the method of the second configuration of the present 
invention may be employed in in vivo therapeutic and prophylactic applications, in vitro 
and in vivo diagnostic applications, in vitro assay and reagent applications, and the like. 
For example antibody molecules may be used in antibody based assay techniques, such as 
20 ELISA techniques, according to methods known to those skilled in the art. 

As alluded to above, the multispecific ligands according to the invention are of use in 
diagnostic, prophylactic and therapeutic procedures. Multispecific antibodies according to 
the invention are of use diagnostically in Western analysis and in situ protein detection by 
25 standard immunohistochemical procedures; for use in these applications, the ligands may 
be labelled in accordance with techniques known to the art. In addition, such antibody 
polypeptides may be used preparatively in affinity chromatography procedures, when 
complexed to a chromatographic support, such as a resin. All such techniques are well 
knowm to one of skill in the art. 

30 

Diagnostic uses of the closed conformation multispecific ligands according to the 
invention include homogenous assays for analytes which exploit the ability of closed 
conformation multispecific ligands to bind two targets in competition, such that two 



WO 2004/003019 PCT/GB2003/002804 

73 

targets cannot bind simultaneously (a closed conformation), or alternatively their ability 
to bind two targets simultaneously (an open conformation). 

A true homogenous immunoassay format has been avidly sought by manufacturers of 
5 diagnostics and research assay systems used in drug discovery and development. The 
main diagnostics markets include human testing in hospitals, doctor's offices and clinics, 
commercial reference laboratories, blood banks, and the home, non-human diagnostics 
(for example food testing, water testing, environmental testing, bio-defence, and 
veterinary testing), and finally research (including drug development; basic research and 
1 o academic research) . 

At present all these markets utilise immunoassay systems that are built around 
chemiluminescent, ELISA, fluorescence or in rare cases radio-immunoassay 
technologies. Each of these assay formats requires a separation step (separating bound 

15 from un-bound reagents). In some cases, several separation steps are required. Adding 
these additional steps adds reagents and automation, takes time, and affects the ultimate 
outcome of the assays. In human diagnostics, the separation step may be automated, 
which masks the problem, but does not remove it. The robotics, additional reagents, 
additional incubation times, and the like add considerable cost and complexity. In drug 

20 development, such as high throughput screening, where literally millions of samples are 
tested at once, with very low levels of test molecule, adding additional separation steps 
can eliminate the ability to perform a screen. However, avoiding the separation creates 
too much noise in the read out. Thus, there is a need for a true homogenous format that 
provides sensitivities at the range obtainable from present assay formats. Advantageously, 

25 an assay possesses fully quantitative read-outs with high sensitivity and a large dynamic 
range. Sensitivity is an important requirement, as is reducing the amount of sample 
required. Both of these features are features that a homogenous system offers. This is 
very important in point of care testing, and in drug development where samples are 
precious. Heterogenous systems, as currently available in the art, require large quantities 

30 of sample and expensive reagents 

Applications for homogenous assays include cancer testing, where the biggest assay is 
that for Prostate Specific Antigen, used in screening men for prostate cancer. Other 



WO 2004/003019 PCT/GB2003/002804 

74 

applications include fertility testing, which provides a series of tests for women 
attempting to conceive including beta-hcg for pregnancy. Tests for infectious diseases, 
including hepatitis, HIV, rubella, and other viruses and microorganisms and sexually 
transmitted diseases. Tests are used by blood banks, especially tests for HIV, hepatitis A, 
5 B, C, non A non B. Therapeutic drug monitoring tests include monitoring levels of 
prescribed drugs in patients for efficacy and to avoid toxicity, for example digoxin for 
arrhythmia, and phenobarbital levels in psychotic cases; theophylline for asthma. 
Diagnostic tests are moreover useful in abused drug testing, such as testing for cocaine, 
marijuana and the like. Metabolic tests are used for measuring thyroid function, anaemia 
10 and other physiological disorders and functions. 

The homogenous immunoassay format is moreover useful in the manufacture of standard 
clinical chemistry assays. The inclusion of immunoassays and chemistry assays on the 
same instrument is highly advantageous in diagnostic testing. Suitable chemical assays 
15 include tests for glucose, cholesterol, potassium, and the like. 

A further major application for homogenous immunoassays is drug discovery and 
development: high throughput screening includes testing combinatorial chemistry 
libraries versus targets in ultra high volume. Signal is detected, and positive groups then 

20 split into smaller groups, and eventually tested in cells and then animals. Homogenous 
assays may be used in all these types of test. In drug development, especially animal 
studies and clinical trials heavy use of immunoassays is made. Homogenous assays 
greatly accelerate and simplify these procedures. Other Applications include food and 
beverage testing: testing meat and other foods for E. coli, salmonella, etc; water testing, 

25 including testing at water plants for all types of contaminants including E. coli; and 
veterinary testing. 

In a broad embodiment, the invention provides a binding assay comprising a detectable 
agent which is bound to a closed conformation multispecific ligand according to the 
30 invention, and whose detectable properties are altered by the binding of an analyte to said 
closed conformation multispecific ligand. Such an assay may be configured in several 
different ways, each exploiting the above properties of closed conformation multispecific 
ligands. 



WO 2004/003019 



75 



PCT/GB2003/002804 



The assay relies on the direct or indirect displacement of an agent by the analyte, resulting 
in a change in the detectable properties of the agent. For example, where the agent is an 
enzyme which is capable of catalysing a reaction which has a detectable end-point, said 
5 enzyme can be bound by the ligand such as to obstruct its active site, thereby inactivating 
the enzyme. The analyte, which is also bound by the closed conformation multispecific 
ligand, displaces the enzyme, rendering it active through freeing of the active site. The 
enzyme is then able to react with a substrate, to give rise to a detectable event. In an 
alternative embodiment, the ligand may bind the enzyme outside of the active site, 
10 influencing the conformation of the enzyme and thus altering its activity. For example, 
the structure of the active site may be constrained by the binding of the ligand, or the 
binding of cofactors necessary for activity may be prevented. 

The physical implementation of the assay may take any form known in the art. For 
15 example, the closed conformation multispecific ligand/enzyme complex may be provided 
on a test strip; the substrate may be provided in a different region of the test strip, and a 
solvent containing the analyte allowed to migrate through the ligand/enzyme complex, 
displacing the enzyme, and carrying it to the substrate region to produce a signal. 
Alternatively, the ligand/enzyme complex may be provided on a test stick or other solid 
20 phase, and dipped into an analyte/substrate solution, releasing enzyme into the solution in 
response to the presence of analyte. 

Since each molecule of analyte potentially releases one enzyme molecule, the assay is 
quantitative, with the strength of the signal generated in a given time being dependent on 
25 the concentration of analyte in the solution. 

Further configurations using the analyte in a closed conformation are possible. For 
example, the closed conformation multispecific ligand may be configured to bind an 
enzyme in an allosteric site, thereby activating the enzyme. In such an embodiment, the 
30 enzyme is active in the absence of analyte. Addition of the analyte displaces the enzyme 
and removes allosteric activation, thus inactivating the enzyme. 



WO 2004/003019 PCT/GB2003/002804 

76 

In the context of the above embodiments which employ enzyme activity as a measure of 
the analyte concentration, activation or inactivation of the enzyme refers to an increase or 
decrease in the activity of the enzyme, measured as the ability of the enzyme to catalyse a 
signal-generating reaction. For example, the enzyme may catalyse the conversion of an 
undetectable substrate to a detectable form thereof. For example, horseradish peroxidase 
is widely used in the art together with chromogenic or chemiluminescent substrates, 
which are available commercially. The level of increase or decrease of the activity of the 
enzyme may between 10% and 100%, such as 20%, 30%, 40%, 50%, 60%, 70%, 80% or 
90%; in the case of an increase in activity, the increase may be more than 100%, i.e. 
200%, 300%, 500% or more, or may not be measurable as a percentage if the baseline 
activity of the inhibited enzyme is undetectable. 

In a further configuration, the closed conformation multispecific ligand may bind the 
substrate of an enzyme/substrate pair, rather than the enzyme. The substrate is therefore 
unavailable to the enzyme until released from the closed conformation multispecific 
ligand through binding of the analyte. The implementations for this configuration are as 
for the configurations which bind enzyme. 

Moreover, the assay may be configured to bind a fluorescent molecule, such as a 
fluorescein or another fluorophore, in a conformation such that the fluorescence is 
quenched on binding to the ligand. In this case, binding of the analyte to the ligand will 
displace the fluorescent molecule, thus producing a signal. Alternatives to fluorescent 
molecules which are useful in the present invention include luminescent agents, such as 
luciferin/luciferase, and chromogenic agents, including agents commonly used in 
immunoassays such as HRP. 

Therapeutic and prophylactic uses of multispecific ligands prepared according to the 
invention involve the administration of ligands according to the invention to a recipient 
mammal, such as a human. Multi-specificity can allow antibodies to bind to multimeric 
antigen with great avidity. Multispecific ligands can allow thecross- linking of two 
antigens, for example in recruiting cytotoxic T-cells to mediate the killing of tumour cell 
lines. 



WO 2004/003019 PCT/GB2003/002804 

77 

Substantially pure ligands or binding proteins thereof, for example dAb monomers, of at 
least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 
99% or more homogeneity is most preferred for pharmaceutical uses, especially when the 
mammal is a human. Once purified, partially or to homogeneity as desired, the ligands 
5 may be used diagnostically or therapeutically (including extracorporeally) or in 
developing and performing assay procedures, immunofluorescent stainings and the like 
(Lefkovite and Pemis, (1979 and 1981) Immunological Methods, Volumes I and II, 
Academic Press, NY). 

10 The ligands or binding proteins thereof, for example dAb monomers, of the present 
invention will typically find use in preventing, suppressing or treating inflammatory 
states, allergic hypersensitivity, cancer, bacterial or viral infection, and autoimmune 
disorders (which include, but are not limited to, Type I diabetes, asthma, multiple 
sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and 

15 myasthenia gravis). 

In the instant application, the term "prevention" involves administration of the protective 
composition prior to the induction of the disease. "Suppression" refers to administration 
of the composition after an inductive event, but prior to the clinical appearance of the 
20 disease. "Treatment" involves administration of the protective composition after disease 
symptoms become manifest. 



Animal model systems which can be used to screen the effectiveness of the antibodies or 
binding proteins thereof in protecting against or treating the disease are available. 

25 Methods for the testing of systemic lupus erythematosus (SLE) in susceptible mice are 
known in the art (Knight et al (1978) J. Exp. Med., 147: 1653; Reinersten et al (1978) 
New Eng. J. Med., 299: 515). Myasthenia Gravis (MG) is tested in SJL/J female mice by 
inducing the disease with soluble AchR protein from another species (Lmdstrom et al 
(1988) Adv. Immunol, 42: 233). Arthritis is induced in a susceptible strain of mice by 

30 injection of Type II collagen (Stuart et al. (1984) Ann. Rev. Immunol, 42: 233). A model 
by which adjuvant arthritis is induced in susceptible rats by injection of mycobacterial 
heat shock protein has been described (Van Eden et al (1988) Nature, 331: 171). 
Thyroiditis is induced in mice by administration of thyroglobulin as described (Maron et 



WO 2004/003019 PCT/GB2003/002804 

78 

al (1980) J. Exp. Med., 152: 1115). Insulin dependent diabetes mellitus (IDDM) occurs 
naturally or can be induced in certain strains of mice such as those described by 
Kanasawa et al (1984) Diabetologia, 27: 113. EAE in mouse and rat serves as a model 
for MS in human. In this model, the demyelinating disease is induced by administration 
of myelin basic protein (see Paterson (1986) Textbook of Immunopathology, Mischer et 
al 9 eds., Grune and Stratton, New York, pp. 179-213; McFarlin et al (1973) Science, 
179: 478: and Satoh et al (1987) J. Immunol, 138: 179). 

Generally, the present ligands will be utilised in purified form together with 
pharmacologically appropriate carriers. Typically, these carriers include aqueous or 
alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or 
buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, 
dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable 
adjuvants, if necessary to keep a polypeptide complex in suspension, maybe chosen from 
thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates. 

Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, 
such as those based on Ringer's dextrose. Preservatives and other additives, such as 
antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack 
(1982) Remington's Pharmaceutical Sciences, 16th Edition). 

The ligands of the present invention may be used as separately administered compositions 
or in conjunction with other agents. These can include various immunotherapeutic drugs, 
such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. 
Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents 
in conjunction with the ligands of the present invention, or even combinations of lignds 
according to the present invention having different specificities, such as ligands selected 
using different target antigens or epitopes, whether or not they are pooled prior to 
administration. 

The route of administration of pharmaceutical compositions according to the invention 
may be any of those commonly known to those of ordinary skill in the art. For therapy, 
including without limitation immunotherapy, the selected ligands thereof of the invention 



WO 2004/003019 PCT/GB2003/002804 

79 

can be administered to any patient in accordance with standard techniques. The 
administration can be by any appropriate mode, including parenterally, intravenously, 
intramuscularly, intraperitoneal^, transdermally, via the pulmonary route, or also, 
appropriately, by direct infusion with a catheter. The dosage and frequency of 
5 administration will depend on the age, sex and condition of the patient, concurrent 
administration of other drugs, counterindications and other parameters to be taken into 
account by the clinician. 

The ligands of this invention can be lyophilised for storage and reconstituted in a suitable 
10 carrier prior to use. This technique has been shown to be effective with conventional 
immunoglobulins and art-known lyophilisation and reconstitution techniques can be 
employed. It will be appreciated by those skilled in the art that lyophilisation and 
reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional 
immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) 
15 and that use levels may have to be adjusted upward to compensate. 

The compositions containing the present ligands or a cocktail thereof can be administered 
for prophylactic and/or therapeutic treatments. In certain therapeutic applications, an 
adequate amount to accomplish at least partial inhibition, suppression, modulation, 

20 killing, or some other measurable parameter, of a population of selected cells is defined as 
a "therapeutically-effective dose". Amounts needed to achieve this dosage will depend 
upon the severity of the disease and the general state of the patient's own immune system, 
but generally range from 0.005 to 5.0 mg of ligand, e.g. antibody, receptor (e.g. a T-cell 
receptor) or binding protein thereof per kilogram of body weight, with doses of 0.05 to 

25 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions 
containing the present ligands or cocktails thereof may also be administered in similar or 
slightly lower dosages. 

Treatment performed using the compositions described herein is considered "effective" if 
30 one or more symptoms is reduced (e.g., by at least 10% or at least one point on a clinical 
assessment scale), relative to such symptoms present before treatment, or relative to such 
symptoms in an individual (human or model animal) not treated with such composition. 
Symptoms will obviously vary depending upon the disease or disorder targeted, but can 



WO 2004/003019 PCT/GB2003/002804 

80 

be measured by an ordinarily skilled clinician or technician. Such symptoms can be 
measured, for example, by monitoring the level of one or more biochemical indicators of 
the disease or disorder (e.g., levels of an enzyme or metabolite correlated with the 
disease, ' affected cell numbers, etc.), by monitoring physical manifestations (e.g., 
5 inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, 
the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory 
Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect 
to bowel function, systemic symptoms, social function and emotional status - score ranges 
from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life 

10 Rheumatoid Arthritis Scale, or other accepted clinical assessment scale as known in the 
field. A sustained (e.g., one day or more, preferably longer) reduction in disease or 
disorder symptoms by at least 10% or by one or more points on a given clinical scale is 
indicative of "effective" treatment. Similarly, prophylaxis performed using a composition 
as described herein is "effective" if the onset or severity of one or more symptoms is 

15 delayed, reduced or abolished relative to such symptoms in a similar individual (human or 
animal model) not treated with the composition. 

A composition containing a ligand or cocktail thereof according to the present invention 
may be utilised in prophylactic and therapeutic settings to aid in the alteration, 

20 inactivation, killing or removal of a select target cell population in a mammal. In addition, 
the selected repertoires of polypeptides described herein may be used extracorporeally or 
in vitro selectively to kill, deplete or otherwise effectively remove a target cell population 
from a heterogeneous collection of cells. Blood from a mammal may be combined 
extracorporeally with the ligands, e.g. antibodies, cell-surface receptors or binding 

25 proteins thereof whereby the undesired cells are killed or otherwise removed from the 
blood for return to the mammal in accordance with standard techniques. 



I: Use of half-life enhanced dual-specific ligands according to the invention 

30 

Dual-specific ligands according to the method of the present invention may be employed 
in in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and 
the like. 



WO 2004/003019 



81 



PCT/GB2003/002804 



Therapeutic and prophylactic uses of dual-specific ligands prepared according to the 
invention involve the administration of ligands according to the invention to a recipient 
mammal, such as a human. Dual specific antibodies according to the invention comprise 
5 at least one specificity for a half-life enhancing molecule; one or more further specificities 
may be directed against target molecules. For example, a dual-specific IgG may be 
specific for four epitopes, one of which is on a half-life enhancing molecule. Dual- 
specificity can allow antibodies to bind to multimeric antigen with great avidity. Dual- 
specific antibodies can allow the cross-linking of two antigens, for example in recruiting 
10 cytotoxic T-cells to mediate the killing of tumour cell lines. 

Substantially pure ligands or binding proteins thereof, such as dAb monomers, of at least 
90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or 
more homogeneity is most preferred for pharmaceutical uses, especially when the 
15 mammal is a human. Once purified, partially or to homogeneity as desired, the ligands 
may be used diagnostically or therapeutically (including extracorporeally) or in 
developing and performing assay procedures, immunofluorescent stainings and the like 
(Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, 
Academic Press, NY). 

20 

The ligands of the present invention will typically find use in preventing, suppressing or 
treating inflammatory states, allergic hypersensitivity, cancer, bacterial or viral infection, 
and autoimmune disorders (which include, but are not limited to, Type I diabetes, 
multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease 
25 and myasthenia gravis). 

In the instant application, the term "prevention" involves administration of the protective 
composition prior to the induction of the disease. "Suppression" refers to administration 
of the composition after an inductive event, but prior to the clinical appearance of the 
30 disease. "Treatment" involves administration of the protective composition after disease 
symptoms become manifest. 



WO 2004/003019 PCT/GB2003/002804 

82 

Animal model systems which can be used to screen the effectiveness of the dual specific 
ligands in protecting against or treating the disease are available. Methods for the testing 
of systemic lupus erythematosus (SLE) in susceptible mice are known in the art (Knight 
et al (1978) J. Exp. Med., 147: 1653; Reinersten et al (1978) New Eng. J. Med, 299: 
5 515). Myasthenia Gravis (MG) is tested in SJL/J female mice by inducing the disease 
with soluble AchR protein from another species (Lindstrom et al (1988) Adv. Immunol, 
42: 233). Arthritis is induced in a susceptible strain of mice by injection of Type II 
collagen (Stuart et al (1984) Ann. Rev. Immunol, 42: 233). A model by which adjuvant 
arthritis is induced in susceptible rats by injection of mycobacterial heat shock protein has 

10 been described (Van Eden et al. (1988) Nature, 331: 171). Thyroiditis is induced in mice 
by administration of thyroglobulin as described (Maron et al (1980) J. Exp. Med., 152: 
1115). Insulin dependent diabetes mellitus (IDDM) occurs naturally or can be induced in 
certain strains of mice such as those described by Kanasawa et al (1984) Diabetologia, 
27: 113. EAE in mouse and rat serves as a model for MS in human. In this model, the 

15 demyelinating disease is induced by administration of myelin basic protein (see Paterson 
(1986) Textbook of Immunopathology, Mischer et al, eds., Grune and Stratton, New 
York, pp. 179-213; McFarlin et al (1973) Science, 179: 478: and Satoh et al (1987) J. 
Immunol, 138: 179). 

20 Dual specific ligands according to the invention and dAb monomers able to bind to 
extracellular targets involved in endocytosis (e.g. Clathrin) enable dual specific ligands to 
be endocytosed, enabling another specificity able to bind to an intracellular target to be 
delivered to an intracellular environment. This strategy requires a dual specific ligand 
with physical properties that enable it to remain functional inside the cell. Alternatively, 

25 if the final destination intracellular compartment is oxidising, a well folding ligand may 
not need to be disulphide free. 

Generally, the present dual specific ligands will be utilised in purified form together with 
pharmacologically appropriate carriers. Typically, these carriers include aqueous or 
30 alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or 
buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, 
dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable 



WO 2004/003019 PCT/GB2003/002804 

83 

adjuvants, if necessary to keep a polypeptide complex in suspension, may be chosen 
from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and 
alginates. 

5 Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishes, 
such as those based on Ringer's dextrose. Preservatives and other additives, such as 
antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack 
(1982) Remington's Pharmaceutical Sciences,, 16th Edition). 



10 The ligands of the present invention may be used as separately administered compositions 
or in conjunction with other agents. These can include various immunotherapeutic drugs, 
such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. 
Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents 
in conjunction with the ligands of the present invention. 

15 

The route of administration of pharmaceutical compositions according to the invention 
may be any of those commonly known to those of ordinary skill in the art. For therapy, 
including without limitation immunotherapy, the ligands of the invention can be 
administered to any patient in accordance with standard techniques. The administration 
20 can be by any appropriate mode, including parenterally, intravenously, intramuscularly, 
intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct 
infusion with a catheter. The dosage and frequency of administration will depend on the 
age, sex and condition of the patient, concurrent administration of other drugs, 
counterindications and other parameters to be taken into account by the clinician. 

25 

The ligands of the invention can be lyophilised for storage and reconstituted in a suitable 
carrier prior to use. This technique has been shown to be effective with conventional 
immunoglobulins and art-known lyophilisation and reconstitution techniques can be 
employed. It will be appreciated by those skilled in the art that lyophilisation and 
30 reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional 
immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) 
and that use levels may have to be adjusted upward to compensate. 



WO 2004/003019 PCT/GB2003/002804 

84 

The compositions containing the present ligands or a cocktail thereof can be 
administered for prophylactic and/or therapeutic treatments. In certain therapeutic 
applications, an adequate amount to accomplish at least partial inhibition, suppression, 
modulation, killing, or some other measurable parameter, of a population of selected cells 
5 is defined as a "therapeutically-effective dose". Amounts needed to achieve this dosage 
will depend upon the severity of the disease and the general state of the patient's own 
immune system, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body 
weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For 
prophylactic applications, compositions containing the present ligands or cocktails thereof 
10 may also be administered in similar or slightly lower dosages. 

A composition containing a ligand according to the present invention may be utilised in 
prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or 
removal of a select target cell population in a mammal 

15 

In addition, the selected repertoires of polypeptides described herein may be used 
extracorporeal^ or in vitro selectively to kill, deplete or otherwise effectively remove a 
target cell population from a heterogeneous collection of cells. Blood from a mammal 
may be combined extracorporeal!/ with the ligands, e.g. antibodies, cell- surface 
20 receptors or binding proteins thereof whereby the undesired cells are killed or otherwise 
removed from the blood for return to the mammal in accordance with standard 
techniques. 

The invention is further described, for the purposes of illustration only, in the following 
25 examples. As used herein, for the purposes of dAb nomenclature, human TNFoc is 
referred to as TAR1 and human TNFoc receptor 1 (p55 receptor) is referred to as TAR2. 

Example 1. Selection of a dual specific scFv antibody (K8) directed against human 
30 serum albumin (HSA) and (3-galactosidase (p -gal) 

This example explains a method for making a dual specific antibody directed against p- 
gal and HSA in which a repertoire of V K variable domains linked to a germline (dummy) 



10 



WO 2004/003019 PCT/GB2003/002804 

85 

V H domain is selected for binding to (3-gal and a repertoire of V H variable domains 
linked to a germline (dummy) V K domain is selected for binding to HSA. The selected 
variable Vh HSA and V K (3-gal domains are then combined and the antibodies selected for 
binding to P-gal and HSA. HSA is a half-life increasing protein found in human blood. 



Four human phage antibody libraries were used in this experiment. 



Library 1 Germline V K /DVT V H 8.46 x 1 0? 

Library 2 Germline V K /NNK V H 9.64 x 10 7 

Library 3 Germline V H /D VT V K 1 .47 x 1 0 8 

Library 4 Germline V H /NNK V K 1 .45 x 1 0 s 



All libraries are based on a single human framework for Vh (V3-23/DP47 and JH4b) and 
V K (012/02/DPK9 and J K 1) with side chain diversity incorporated in complementarity 

15 determining regions (CDR2 and CDR3). 



Library 1 and Library 2 contain a dummy V K sequence, whereas the sequence of Vh is 

diversified at positions H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97 and H98 
(DVT or NNK encoded, respectively) (Figure 1). Library 3 and Library 4 contain a 
20 dummy Vh sequence, whereas the sequence of V K is diversified at positions L50, L53, 

L91, L92, L93, L94 and L96 (DVT or NNK encoded, respectively) (Figure 1). The 
libraries are in phagemid pIT2/ScFv format (Figure 2) and have been preselected for 
binding to generic ligands, Protein A and Protein L, so that the majority of clones in the 
unselected libraries are functional. The sizes of the libraries shown above correspond to 
25 the sizes after preselection. Library 1 and Library 2 were mixed prior to selections on 
antigen to yield a single Vn/dummy V K library and Library 3 and Library 4 were mixed 

to form a single V K /dummy Vh library. 



Three rounds of selections were performed on (3-gal using V K /dummy V H library and 
30 three rounds of selections were performed on HSA using V H /dummy V K library. In the 



WO 2004/003019 PCT/GB2003/002804 

86 

case of P-gal the phage titres went up from 1.1 x 10 6 in the first round to 2.0 x 10 8 in the 

third round. In the case of HSA the phage titres went up from 2 x 10 4 in the first round to 

1.4 x 10 9 in the third round. The selections were performed as described by Griffith et aL, 
(1993), except that KM13 helper phage (which contains a pill protein with a protease 
5 cleavage site between the D2 and D3 domains) was used and phage were eluted with 1 
mg/ml trypsin in PBS. The addition of trypsin cleaves the pill proteins derived from the 
helper phage (but not those from the phagemid) and elutes bound scFv-phage fusions by 
cleavage in the c-myc tag (Figure 2), thereby providing a further enrichment for phages 
expressing functional scFvs and a corresponding reduction in background (Kristensen & 
10 Winter, Folding & Design 3: 321-328, Jul 9, 1998). Selections were performed using 
immunotubes coated with either HSA or p-gal at 100p,g/ml concentration. 

To check for binding, 24 colonies from the third round of each selection were screened by 
monoclonal phage ELISA. Phage particles were produced as described by Harrison et al. 9 
15 Methods Enzymol. 1996;267:83-109. 96-well ELISA plates were coated with lOOjul of 
HSA or P-gal at 10|j,g/ml concentration in PBS overnight at 4°C. A standard ELISA 
protocol was followed (Hoogenboom et al. 9 1991) using detection of bound phage with 
anti-M13-HRP conjugate. A selection of clones gave ELISA signals of greater than 1.0 
with 50|al supernatant. 

20 

Next, DNA preps were made from V H /dummy V K library selected on HSA and from 

V K /dummy V H library selected on p-gal using the QIAprep Spin Miniprep kit (Qiagen). 

To access most of the diversity, DNA preps were made from each of the three rounds of 
selections and then pulled together for each of the antigens. DNA preps were then 
25 digested with Sali/NotT overnight at 37°C. Following gel purification of the fragments, 
V K chains from the V K /dummy V H library selected on P-gal were ligated in place of a 

dummy V K chain of the V H /dummy V K library selected on HSA creating a library of 3.3 
x 10^ clones. 

30 This library was then either selected on HSA (first round) and p-gal (second round), 
HSA/p-gal selection, or on p-gal (first round) and HSA (second round), P-gal/HSA 



WO 2004/003019 PCT/GB2003/002804 

87 

selection. Selections were performed as described above. In each case after the second 
round 48 clones were tested for binding to HSA and P-gal by the monoclonal phage 
ELISA (as described above) and by ELISA of the soluble scFv fragments. Soluble 
antibody fragments were produced as described by Harrison et aL, (1996), and standard 
5 ELISA protocol was followed Hoogenboom et al. (1991) Nucleic Acids Res., 19: 4133, 
except that 2% Tween/PBS was used as a blocking buffer and bound scFvs were detected 
with Protein L-HRP. Three clones (E4, E5 and E8) from the HSA/p-gal selection and two 
clones (K8 and K10) from the (3-gal/HSA selection were able to bind both antigens. scFvs 
from these clones were PCR amplified and sequenced as described by Ignatovich et aL 9 
10 (1999) J Mol Biol 1999 Nov 26;294(2):457-65, using the primers LMB3 and pHENseq. 
Sequence analysis revealed that all clones were identical. Therefore, only one clone 
encoding a dual specific antibody (K8) was chosen for further work (Figure 3). 



15 Example 2. Characterisation of the binding properties of the K8 antibody. 

Firstly, the binding properties of the K8 antibody were characterised by the monoclonal 
phage ELISA. A 96-well plate was coated with lOOjul of HSA and p-gal alongside with 
alkaline phosphatase (APS), bovine serum albumin (BSA), peanut agglutinin, lysozyme 

20 and cytochrome c (to check for cross-reactivity) at 10|Lig/ml concentration in PBS 
overnight at 4°C. The phagemid from K8 clone was rescued with KM13 as described by 
Harrison et al. 9 (1996) and the supernatant (50|lx1) containing phage assayed directly. A 
standard ELISA protocol was followed (Hoogenboom et aL 9 1991) using detection of 
bound phage with anti-M13-HRP conjugate. The dual specific K8 antibody was found to 

25 bind to HSA and p-gal when displayed on the surface of the phage with absorbance 
signals greater than 1.0 (Figure 4). Strong binding to BSA was also observed (Figure 4). 
Since HSA and BSA are 76% homologous on the amino acid level, it is not surprising 
that K8 antibody recognised both of these structurally related proteins. No cross-reactivity 
with other proteins was detected (Figure 4). 

30 

Secondly, the binding properties of the K8 antibody were tested in a soluble scFv ELISA. 
Production of the soluble scFv fragment was induced by IPTG as described by Harrison 
et al. 9 (1996). To determine the expression levels of K8 scFv, the soluble antibody 



WO 2004/003019 PCT/GB2003/002804 

88 

fragments were purified from the supernatant of 50ml inductions using Protein A- 
Sepharose columns as described by Harlow and Lane, Antibodies: a Laboratory Manual, 
(1988) Cold Spring Harbor. OD28O was ^en measured and the protein concentration 

calculated as described by Sambrook et aL, (1989). K8 scFv was produced in supernatant 
5 at 19mg/l. 

A soluble scFv ELISA was then performed using known concentrations of the K8 
antibody fragment. A 96-well plate was coated with 100|ul of HSA, BSA and p-gal at 
lOjag/ml and lOOjul of Protein A at 1 \xg/ml concentration. 50jli1 of the serial dilutions of 
10 the K8 scFv was applied and the bound antibody fragments were detected with Protein L- 
HRP. ELISA results confirmed the dual specific nature of the K8 antibody (Figure 5). 

To confirm that binding to (3-gal is determined by the V K domain and binding to 

HSA/BSA by the Vh domain of the K8 scFv antibody, the V K domain was cut out from 

15 K8 scFv DNA by Sali/Not/ digestion and ligated into a SalZ/NotT digested pIT2 vector 
containing dummy V H chain (Figures 1 and 2). Binding characteristics of the resulting 
clone K8V K /dummy Vh were analysed by soluble scFv ELISA. Production of the soluble 

scFv fragments was induced by IPTG as described by Harrison et al. 9 (1996) and the 
supernatant (50|lx) containing scFvs assayed directly. Soluble scFv ELISA was performed 
20 as described in Example 1 and the bound scFvs were detected with Protein L-HRP. The 
ELISA results revealed that this clone was still able to bind p-gal, whereas binding to 
BSA was abolished (Figure 6). 

Example 3. Selection of single Vjj domain antibodies antigens A and B and single 
25 V K domain antibodies directed against antigens C and D. 

This example describes a method for making single Vjj domain antibodies directed 

against antigens A and B and single V K domain antibodies directed against antigens C 

and D by selecting repertoires of virgin single antibody variable domains for binding to 
30 these antigens in the absence of the complementary variable domains. 



WO 2004/003019 PCT/GB2003/002804 

89 

Selections and characterisation of the binding clones is performed as described 
previously (see Example 5, PCT/GB 02/003014). Four clones are chosen for further 
work: 

VHl-Anti AV H 
VH2-AntiB V H 
VK1 - Anti C V K 
VK2 — Anti D V K 

The procedures described above in Examples 1-3 may be used, in a similar manner as that 
described, to produce dimer molecules comprising combinations of V H domains (i.e., V H - 
Vh ligands) and cominations of Vl domains (V L -V L ligands). 

Example 4. Creation and characterisation of the dual specific ScFv antibodies 
(VH1/VH2 directed against antigens A and B and VK1 /VK2 directed against 
antigens C and D). 

This example demonstrates that dual specific ScFv antibodies (VH1/VH2 directed against 
antigens A and B and VK1/VK2 directed against antigens C and D) could be created by 
combining V K and V H single domains selected against respective antigens in a ScFv 
vector. 

To create dual specific antibody VH1 /VH2, VH1 single domain is excised from variable 
domain vector 1 (Figure 7) by NcoVXhdl digestion and ligated into NcoVXlioI digested 
variable domain vector 2 (Figure 7) to create VH1/ variable domain vector 2. VH2 single 
domain is PGR amplified from variable domain vector 1 using primers to introduce Sail 
restriction site to the 5' end and NotI restriction site to the 3 5 end. The PCR product is 
then digested with SaWNotl and ligated into SaWNotl digested VH1/ variable domain 
vector 2 to create VH1/VH2/ variable domain vector 2. 

VK1/VK2/ variable domain vector 2 is created in a similar way. The dual specific nature 
of the produced VH1/VH2 ScFv and VK1/VK2 ScFv is tested in a soluble ScFv ELISA 



WO 2004/003019 PCT/GB2003/002804 

90 

as described previously (see Example 6, PCT/GB 02/003014). Competition ELISA is 
performed as described previously (see Example 8, PCT/GB 02/003014). 

Possible outcomes: 

-VH1/VH2 ScFv is able to bind antigens A and B simultaneously 

-VK1/VK2 ScFv is able to bind antigens C and D simultaneously 

-VH1/VH2 ScFv binding is competitive (when bound to antigen A, VH1/VH2 ScFv 

cannot bind to antigen B) 

-VK1/VK2 ScFv binding is competitive (when bound to antigen C, VK1/VK2 ScFv 
cannot bind to antigen D) 

Example 5. Construction of dual specific VH1/VH2 Fab and VK1/VK2 Fab and 
analysis of their binding properties. 

To create VH1/VH2 Fab, VH1 single domain is ligated into NcoVXhol digested CH 
vector (Figure 8) to create VH1/CH and VH2 single domain is ligated into SaWNotl 
digested CK vector (Figure 9) to create VH2/CK. Plasmid DNA from VH1/CH and 
VH2/CK is used to co-transform competent E. coli cells as described previously (see 
Example 8, PCT/GB02/003014). 

The clone containing VH1/CH and VH2/CK plasmids is then induced by IPTG to 
produce soluble VH1/VH2 Fab as described previously (see Example 8, PCT/GB 
02/003014). 

VK1/VK2 Fab is produced in a similar way. 

Binding properties of the produced Fabs are tested by competition ELISA as described 
previously (see Example 8, PCT/GB 02/003014). 

Possible outcomes: 

-VH1/VH2 Fab is able to bind antigens A and B simultaneously 
-VK1/VK2 Fab is able to bind antigens C and D simultaneously 



WO 2004/003019 PCT/GB2003/002804 

91 

-VH1/VH2 Fab binding is competitive (when bound to antigen A, VH1/VH2 Fab cannot 
bind to antigen B) 

-VK1/VK2 Fab binding is competitive (when bound to antigen C, VK1/VK2 Fab cannot 
bind to antigen D) 

5 

Example 6 

Chelating dAb Dimers 
10 Summary 

VH and VK homo-dimers are created in a dAb-linker-dAb format using flexible 
polypeptide linkers. Vectors were created in the dAb linker-dAb format containing 
glycine-serine linkers of different lengths 3U:(Gly4Ser) 3 , 5U:(Gly4Ser) 5 , 7U:(Gly4Ser) 7 . 
Dimer libraries were created using guiding dAbs upstream of the linker: TAR1-5 (VK), 
15 TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) and a library of corresponding second 
dAbs after the linker. Using this method, novel dimeric dAbs were selected. The effect of 
dimerisation on antigen binding was determined by ELISA and BIAcore studies and in 
cell neutralisation and receptor binding assays. Dimerisation of both TAR1-5 and TAR1- 
27 resulted in significant improvement in binding affinity and neutralisation levels. 

20 

1.0 Methods 

1.1 Library generation 
1.1.1 Vectors 

pEDA3U, pEDA5U and pEDA7U vectors were designed to introduce different linker 
25 lengths compatible with the dAb-linker-dAb format. For pEDA3U, sense and anti-sense 
73 -base pair oligo linkers were annealed using a slow annealing program (95°C-5mins, 
80°C-10mins, 70°C-15mins 5 56°C-15mins ? 42°C until use) in buffer containing 
O.lMNaCl, lOmM Tris-HCl pH7.4 and cloned using the XJ10I and Notl restriction sites. 
The linkers encompassed 3 (Gly4Ser) units and a staffer region housed between Sail and 
30 Notl cloning sites (scheme 1). In order to reduce the possibility of monomelic dAbs 
being selected for by phage display, the stuffer region was designed to include 3 stop 
codons, a Sacl restriction site and a frame shift mutation to put the region out of frame 
when no second dAb was present. For pEDASU and 7U due to the length of the linkers 



WO 2004/003019 PCT/GB2003/002804 

92 

required, overlapping oligo-linkers were designed for each vector, annealed and 
elongated using Klenow. The fragment was then purified and digested using the 
appropriate enzymes before cloning using ths Xhol and Notl restriction sites. 



Ncol 



Xhol 



Linker: 
3U 



5U 



Sail 



71) 



r 



Notl 



Stuffer 1 



Staffer 2 



Scheme 1 



1.1.2 Library preparation 

The N~terminal V gene corresponding to the guiding dAb was cloned upstream of the 
10 linker using Ncol and Xhol restriction sites. VH genes have existing compatible sites, 
however cloning VK genes required the introduction of suitable restriction sites. This was 
achieved by using modifying PCR primers (VK-DLIBF: 5' cggccatggcgtcaacggacat; 
VKXholR: 5' atgtgcgctcgagcgtttgattt 3') in 30 cycles of PCR amplification using a 2:1 
mixture of SuperTaq (HTBiotechnology Ltd)and pfu turbo (Stratagene). This maintained 
15 the Ncol site at the 5' end while destroying the adjacent Sail site and introduced the 
Xhol site at the 3' end. 5 guiding dAbs were cloned into each of the 3 dimer vectors: 
TAR1-5 (VK), TAR1-27(VK), TAR2-5(VH), TAR2-6(VK) and TAR2-7(VK). All 
constructs were verified by sequence analysis. 



20 Having cloned the guiding dAbs upstream of the linker in each of the vectors (pEDA3U, 
5U and 7U): TAR1-5 (VK), TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) a library of 
corresponding second dAbs were cloned after the linker. To achieve this, the 
complimentary dAb libraries were PCR amplified from phage recovered from round 1 
selections of either a VK library against Human TNFoc (at approximately 1 x 10 6 diversity 

25 after round 1) when TAR1-5 or TAR1-27 are the guiding dAbs, or a VH or VK library 
against human p55 TNP receptor (both at approximately 1 x 10 5 diversity after round 1) 
when TAR2-5 or TAR2-6 respectively are the guiding dAbs. For VK libraries PCR 
amplification was conducted using primers in 30 cycles of PCR amplification using a 2:1 
mixture of SuperTaq and pfu turbo. VH libraries were PCR amplified using primers in 



WO 2004/003019 PCT/GB2003/002804 

93 

order to introduce a Sail restriction site at the 5' end of the gene. The dAb library PCRs 
were digested with the appropriate restriction enzymes, ligated into the corresponding 
vectors down stream of the linker, using Sall/Notl restriction sites and electroporated 
into freshly prepared competent TGI cells. 

5 

The titres achieved for each library are as follows: 
TAR1-5: pEDA3U = 4xl0 8 , pEDA5U = 8xl0 7 , pEDA7U = 1x10 s 
TAR1-27: pEDA3U = 6.2xl0 8 , pEDA5U = 1x10 s , pEDA7U = lxlO 9 
TAR2h-5: pEDA3U = 4xl0 7 , pEDA5U = 2 x 10 8 , pEDA7U = 8xl0 7 
10 TAR2h-6: pEDASU = 7.4xl0 8 , pEDASU = 1.2 x 10 8 , pEDA7U - 2.2xl0 8 



1.2 Selections 
1.2.1 TNFa 

Selections were conducted using human TNFa passively coated on immunotubes. 

15 Briefly, Immunotubes are coated overnight with 1 -4mls of the required antigen. The 
immunotubes were then washed 3 times with PBS and blocked with 2%milk powder in 
PBS for l-2hrs and washed a further 3 times with PBS. The phage solution is diluted in 
2%milk powder in PBS and incubated at room temperature for 2hrs. The tubes are then 
washed with PBS and the phage eluted with lmg/ml trypsin-PBS. Three selection 

20 strategies were investigated for the TAR 1-5 dimer libraries. The first round selections 
were carried out in immunotubes using human TNFa coated at 1 jag/ml or 20|ug/ml with 
20 washes in PBS 0.1%Tween. TGI cells are infected with the eluted phage and the titres 
are determined (eg, Marks et al J Mol Biol. 1991 Dec 5;222(3):581-97, Richmann et al 
Biochemistry. 1993 Aug 31;32(34):8848-55). 

25 

The titres recovered were: 

pEDA3U = 2.8xl0 7 (l^g/ml TNF) 1.5x10 s (20]Lig/mlTNF), 
pEDA5U = 1.8xl0 7 (ljag/ml TNF), 1.6x10 s (20|ag/ml TNF) 
pEDA7U = 8xl0 6 (lug/ml TNF), 7xl0 7 (20p,g/ml TNF). 

30 

The second round selections were carried out using 3 different methods. 

1. In immunotubes, 20 washes with overnight incubation followed by a further 10 
washes. 



WO 2004/003019 PCT/GB2003/002804 

94 

2. In immunotubes, 20 washes followed by lhr incubation at RT in wash buffer 

with (ljj,g/ml TNFa) and 10 further washes. 
3. Selection on streptavidin beads using 33 pmoles biotinylated human TNFa 
(Henderikx et aL, 2002, Selection of antibodies against biotinylated antigens. 
Antibody Phage Display : Methods and protocols, Ed. O'Brien and Atkin, Humana 
Press). Single clones from round 2 selections were picked into 96 well plates and 
crude supernatant preps were made in 2ml 96 well plate format. 





Round 1 


Round 2 


Round 2 


Round 2 




Human 


selection 


selection 


selection 




TNFaimmuno 


method 1 


method 2 


method 3 




tube coating 










concentration 








pEDA3U 


lu.g/ml 


1 x 10 y 


1.8 x 10 y 


2.4 x 10 1U 


pEDA3U 


20(j.g/ml 


6x 10 y 


1.8 x 10 10 


8.5 x 10 iO 


pEDA5U 


l|j,g/ml 


9x 10* 


1.4 x 10 y 


2.8 x 10 10 


pEDA5U 


20|j,g/ml 


9.5 x 10 y 


8.5 x 10 9 


2.8 x 10 10 


pEDA7U 


lu-g/ml 


7.8 x 10 s 


1.6 x 10* 


4x 10 1U 


pEDA7U 


20u.g/ml 


1 x 10 1U 


8x 10 9 


1.5 x 10 10 



For TAR1-27, selections were carried out as described previously with the following 
modifications. The first round selections were carried out in immunotubes using human 
TNFa coated at l|Lxg/ml or 20jj.g/ml with 20 washes in PBS 0.1%Tween. The second 
round selections were carried out in immunotubes using 20 washes with overnight 
incubation followed by a further 20 washes. Single clones from round 2 selections were 
picked into 96 well plates and crude supernatant preps were made in 2ml 96 well plate 
format. 



TAR1-27 titres are as follows: 



WO 2004/003019 



PCT/GB2003/002804 



95 





T-Ti Tm qti 

jn uiiid.il 

TTsTF' a 1 Tn m 1 1 n r\ti i V» 
X1NJL L^llillll U.11L/LU.UC 

coatine cone 


IVOUIIU- 1 


XvUUllU. Z, 


pEDA3U 


lug/ml 


4x 10 y 


6x 10 y 


pEDA3U 


20 jag/ml 


5x 10 y 


4.4 x 10 10 


pEDA5U 


l|ng/ml 


1.5 x 10 y 


1.9 x 10 i0 


pEDA5U 


20|iig/ml 


3.4 x 10 y 


3.5 x 10 10 


pEDA7U 


l|ag/ml 


2.6 x 10 y 


5x 10 y 


pEDA7U 


20p,g/ml 


7xlO y 


1.4 x 10 i0 



1.2.2 TNF RECEPTOR 1 (p55 RECEPTOR; TAR2) 

Selections were conducted as described previously for the TAR2h-5 libraries only. 3 
rounds of selections were carried out in immunotubes using either l|j.g/ml human p55 
5 TNF receptor or 10|Lig/ml human p55 TNF receptor with 20 washes in PBS 0.1%Tween 
with overnight incubation followed by a further 20 washes. Single clones from round 2 
and 3 selections were picked into 96 well plates and crude supernatant preps were made 
in 2ml 96 well plate format. 

10 TAR2h-5 titres are as follows: 





Round 1 human 

p55 TNF 

receptor 

immunotube 

coating 

concentration 


Round 1 


Round 2 


Round 3 


pEDA3U 


l[j.g/ml 


2.4 x 10 6 


1.2 x 10 7 


1.9 x 10 y 


pEDA3U 


10u.g/ml 


3.1 x 10 7 


7 x 10' 


1 x 10 y 


pEDA5U 


l|j.g/ml 


2.5 x 10 & 


1.1 x 10 7 


5.7 x 10 s 


pEDA5U 


10u.g/ml 


3.7 x 10 7 


2.3 x 10 K 


2.9 x 10 y 


pEDA7U 


lM.g/ml 


1.3 x 10 6 


1.3 x 10' 


1.4 x 10 y 


pEDA7U 


10|j.g/ml 


1.6 x 10 7 


1.9 x 10 7 


3x 10 1U 



1.3 Screening 

Single clones from round 2 or 3 selections were picked from each of the 3U 5 5U and 7U 
15 libraries from the different selections methods, where appropriate. Clones were grown in 
2xTY with 100(j,g/ml ampicillin and 1% glucose overnight at 37°C. A 1/100 dilution of 
this culture was inoculated into 2mls of 2xTY with lOOjLig/ml ampicillin and 0.1% 
glucose in 2ml, 96 well plate format and grown at 37°C shaking until OD600 was 



WO 2004/003019 PCT/GB2003/002804 

96 

approximately 0.9. The culture was then induced with ImM IPTG overnight at 30°C. 
The supernatants were clarified by centrifugation at 4000rpm for 15 mins in a sorval plate 
centrifuge. The supernatant preps the used for initial screening. 

1.3.1 ELISA 

Binding activity of dimeric recombinant proteins was compared to monomer by Protein 
A/L ELISA or by antigen ELISA. Briefly, a 96 well plate is coated with antigen or 
Protein A/L overnight at 4°C. The plate washed with 0.05% Tween-PBS, blocked for 2hrs 
with 2% Tween-PBS. The sample is added to the plate incubated for 1 hr at room 
temperature. The plate is washed and incubated with the secondary reagent for lhr at 
room temperature. The plate is washed and developed with TMB substrate. Protein A/L- 
HRP or fridia-HRP was used as a secondary reagent. For antigen ELISAs, the antigen 
concentrations used were ljug/ml in PBS for Human TNFa and human THF receptor 1. 
Due to the presence of the guiding dAb in most cases dimers gave a positive ELISA 
signal therefore off rate determination was examined by BIAcore. 

1.3.2 BIAcore 

BIAcore analysys was conducted for TAR1-5 and TAR2h-5 clones. For screening, 
Human TNFotwas coupled to a CMS chip at high density (approximately 10000 RUs). 
50 jlxI of Human TNFoc(50 |Lig/ml) was coupled to the chip at 5|al/min in acetate buffer - 
pH5.5. Regeneration of the chip following analysis using the standard methods is not 
possible due to the instability of Human TNFa, therefore after each sample was analysed, 
the chip was washed for 1 Omins with buffer. 

For TAR1-5, clones supernatants from the round 2 selection were screened by BIAcore. 
48 clones were screened from each of the 3U, 5U and 7U libraries obtained using the 
following selection methods: 

Rl: lju,g/ml human TNFa immunotube, R2 1 jag/ml human TNFa immunotube, overnight 
wash. 

Rl: 20/ig/ml human TNFa immunotube, R2 20jng/ml human TNFa immunotube, 
overnight wash. 

Rl: l/ig/ml human TNFa immunotube, R2 33 pmoles biotinylated human TNFa on 
beads. 

Rl : 20jtig/ml human TNFa immunotube, R2 33 pmoles biotinylated human TNFa beads. 



WO 2004/003019 



97 



PCT/GB2003/002804 



For screening, human p55 TNF receptor was coupled to a CM5 chip at high density 
(approximately 4000 RUs). 100 jllI of human p55 TNF receptor (10 )ng/ml) was coupled 
to the chip at 5pl/min in acetate buffer - pH5.5. Standard regeneration conditions were 
5 examined ( glycine pH2 or pH3) but in each case antigen was removed from the surface 
of the chip therefore as with TNFa, therefore after each sample was analysed, the chip 
was washed for 1 Omins with buffer. 

For TAR2-5, clones supernatants from the round 2 selection were screened. 
48 clones were screened from each of the 3U, 5U and 7U libraries, using the following 
1 0 selection methods : 

Rl: 1/xg/ml human p55 TNF receptor immunotube, R2 1/xg/ml human p55 TNF receptor 
immunotube, overnight wash. 

Rl: 1 0/xg/ml human p55 TNF receptor immunotube, R2 10/xg/ml human p55 TNF 
receptor immunotube, overnight wash. 

15 

1.3.3 Receptor and Cell Assays 

The ability of the dimers to neutralise in the receptor assay was conducted as follows: 
Receptor binding 

20 Anti-TNF dAbs were tested for the ability to inhibit the binding of TNF to recombinant 
TNF receptor 1 (p55). Briefly, Maxisorp plates were incubated overnight with 30mg/ml 
anti-human Fc mouse monoclonal antibody (Zymed, San Francisco, USA). The wells 
were washed with phosphate buffered saline (PBS) containing 0.05% Tween-20 and then 
blocked with 1% BSA in PBS before being incubated with lOOng/ml TNF receptor 1 Fc 

25 fusion protein (R&D Systems, Minneapolis, USA). Anti-TNF dAb was mixed with TNF 
which was added to the washed wells at a final concentration of lOng/ml. TNF binding 
was detected with 0.2mg/ml biotinylated anti-TNF antibody (HyCult biotechnology, 
Uben, Netherlands) followed by 1 in 500 dilution of horse radish peroxidase labelled 
streptavidin (Amersham Biosciences, UK) and then incubation with TMB substrate (KPL, 

30 Gaithersburg, USA). The reaction was stopped by the addition of HC1 and the absorb ance 
was read at 450nm. Anti-TNF dAb activity lead to a decrease in TNF binding and 
therefore a decrease in absorbance compared with the TNF only control. 



WO 2004/003019 PCT/GB2003/002804 

98 

L929 Cytotoxicity Assay 

Anti-TNF dAbs were also tested for the ability to neutralise the cytotoxic activity of TNF 
on mouse L929 fibroblasts (Evans, T. (2000) Molecular Biotechnology 15, 243-248). 
Briefly, L929 cells plated in microtitre plates were incubated overnight with anti-TNF 
dAb, lOOpg/ml TNF and lmg/ml actinomycin D (Sigma, Poole, UK). Cell viability was 
measured by reading absorbance at 490nm following an incubation with [3-(4,5- 
dimethylthiazol-2-yl)-5-(3-carbboxymethoxyphenyl)-2-(4-sulfophenyl)-2H 
(Promega, Madison, USA). Anti-TNF dAb activity lead to a decrease in TNF cytotoxicity 
and therefore an increase in absorbance compared with the TNF only control. 

In the initial screen, supernatants prepared for BIAcore analysis, described above, were 
also used in the receptor assay. Further analysis of selected dimers was also conducted in 
the receptor and cell assays using purified proteins. 

HeLa IL-8 assay 

Anti-TNFRl or anti-TNF alpha dAbs were tested for the ability to neutralise the 
induction of IL-8 secretion by TNF in HeLa cells (method adapted from that of Akeson, 
L. et al (1996) Journal of Biological Chemistry 271, 30517-30523, describing the 
induction of IL-8 by IL-1 in HUVEC; here we look at induction by human TNF alpha and 
we use HeLa cells instead of the HUVEC cell line). Briefly, HeLa cells plated in 
microtitre plates were incubated overnight with dAb and 300pg/ml TNF. Post incubation 
the supernatant was aspirated off the cells and IL-8 concentration measured via a 
sandwich ELISA (R&D Systems). Anti-TNFRl dAb activity lead to a decrease in IL-8 
secretion into the supernatant compared with the TNF only control. 

The L929 assay is used throughout the following experiments; however, the use of the 
HeLa IL-8 assay is preferred to measure anti-TNF receptor 1 (p55) ligands; the presence 
of mouse p55 in the L929 assay poses certain limitations in its use. 

1.4 Sequence analysis 

Dimers that proved to have interesting properties in the BIAcore and the receptor assay 
screens were sequenced. Sequences are detailed in the sequence listing. 



WO 2004/003019 PCT/GB2003/002804 

99 

1 .5 Formatting 

1.5.1 TAR1-5-19 dimers 

The TAR1-5 dimers that were shown to have good neutralisation properties were re- 
formatted and analysed in the cell and receptor assays. The TAR 1-5 guiding dab was 
5 substituted with the affinity matured clone TAR1-5-19. To achieve this TAR 1-5 was 
cloned out of the individual dimer pair and substituted with TAR1-5-19 that had been 
amplified by PCR. In addition, TAR 1-5- 19 homodimers were also constructed in the 3U, 
5U and 7U vectors. The N terminal copy of the gene was amplified by PCR and cloned as 
described above and the C-terminal gene fragment was cloned using existing Sail and 
10 Notl restriction sites. 

1.5.2 Mutagenesis 

The amber stop codon present in dAb2, one of the C-terminal dAbs in the TAR 1-5 dimer 
pairs was mutated to a glutamine by site-directed mutagenesis. 

15 

1.5.3 Fabs 

The dimers containing TAR 1-5 or TAR1-5-19 were re-formatted into Fab expression 
vectors. dAbs were cloned into expression vectors containing either the CK or CH genes 
using Sfil and Notl restriction sites and verified by sequence analysis. The CK vector is 
20 derived from a pUC based ampicillin resistant vector and the CH vector is derived from a 
pACYC chloramphenicol resistant vector. For Fab expression the dAb-CH and dAb-CK 
constructs were co-transformed into HB2151 cells and grown in 2xTY containing 0.1% 
glucose, 100ju,g/ml ampicillin and lO/jg/ml chloramphenicol. 

25 1.5.3 Hinge dimerisation 

Dimerisation of dAbs via cystine bond formation was examined. A short sequence of 
amino acids EPKS GDKTHTCPP CP a modified form of the human IgGCl hinge was 
engineered at the C terminal region on the dAb. An oligo linker encoding for this 
sequence was synthesised and annealed, as described previously. The linker was cloned 
30 into the pEDA vector containing TAR1-5-19 using Xhol and Notl restriction sites. 
Dimerisation occurs in situ in the periplasm. 

1.6 Expression and purification 



WO 2004/003019 PCT/GB2003/002804 

100 

1.6.1 Expression 

Supernatants were prepared in the 2ml, 96-well plate format for the initial screening as 
described previously. Following the initial screening process selected dimers were 
analysed further. Dirtier constructs were expressed in TOP10F' or HB2151 cells as 
supernatants. Briefly, an individual colony from a freshly streaked plate was grown 
overnight at 37°C in 2xTY with lOOpg/ml ampicillin and 1% glucose. A 1/100 dilution 
of this culture was inoculated into 2xTY with 100p.g/ml ampicillin and 0.1% glucose and 
grown at 37°C shaking until OD600 was approximately 0.9. The culture was then induced 
with ImM IPTG overnight at 30°C. The cells were removed by centrifugation and the 
supernatant purified with protein A or L agarose. 

Fab and cysteine hinge dimers were expressed as periplasmic proteins in HB2152 cells. 
A 1/100 dilution of an overnight culture was inoculated into 2xTY with 0.1% glucose 
and the appropriate antibiotics and grown at 30°C shaking until OD600 was 
approximately 0.9. The culture was then induced with ImM IPTG for 3-4 hours at 25°C. 
The cells were harvested by centrifugation and the pellet resuspended in periplasmic 
preparation buffer (30mM Tris-HCl pH8.0, ImM EDTA, 20% sucrose). Following 
centrifugation the supernatant was retained and the pellet resuspended in 5mM MgS04. 
The supernatant was harvested again by centrifugation, pooled and purified. 

1.6.2 Protein A/L purification 

Optimisation of the purification of dimer proteins from Protein L agarose (Affitech, 
Norway) or Protein A agarose (Sigma, UK) was examined. Protein was eluted by batch or 
by column elution using a peristaltic pump. Three buffers were examined 0.1M 
Phosphate-citrate buffer pH2.6, 0.2M Glycine pH2.5 and 0.1M Glycine pH2.5. The 
optimal condition was determined to be under peristaltic pump conditions using 0.1M 
Glycine pH2.5 over 10 column volumes. Purification from protein A was conducted 
peristaltic pump conditions using 0.1M Glycine pH2.5. 

1.6.3 FPLC purification 

Further purification was carried out by FPLC analysis on the AKTA Explorer 1 00 system 
(Amersham Biosciences Ltd). TAR1-5 and TAR1-5-19 dimers were fractionated by 
cation exchange chromatography (1ml Resource S — Amersham Biosciences Ltd) eluted 
with a 0-1M NaCl gradient in 50mM acetate buffer pH4. Hinge dimers were purified by 



WO 2004/003019 



PCT/GB2003/002804 



101 

ion exchange (1ml Resource Q Amersham Biosciences Ltd) eluted with a 0-1M NaCl 
gradient in 25mMTris HC1 pH 8.0. Fabs were purified by size exclusion chromatography 
using a superose 12 (Amersham Biosciences Ltd ) column run at a flow rate of 0.5ml/min 
in PBS with 0.05% tween. Following purification samples were concentrated using 
vivaspin 5K cut off concentrators (Vivascience Ltd). 

2.0 Results 

2.1 TAR1-5 dimers 

6 x 96 clones were picked from the round 2 selection encompassing all the libraries and 
selection conditions. Supernatant preps were made and assayed by antigen and Protein L 
ELISA, BIAcore and in the receptor assays. In ELISAs, positive binding clones were 
identified from each selection method and were distributed between 3U ? 5U and 7U 
libraries. However, as the guiding dAb is always present it was not possible to 
discriminate between high and low affinity binders by this method therefore BIAcore 
analysis was conducted. 

BIAcore analysis was conducted using the 2ml supematants. BIAcore analysis revealed 
that the dimer Koff rates were vastly improved compared to monomelic TAR 1-5. 
Monomer Koff rate was in the range of 1 0^M compared with dimer Koff rates which 
were in the range of 10" 3 - 10~ 4 M. 16 clones that appeared to have very slow off rates 
were selected, these came from the 3U, 5U and 7U libraries and were sequenced. In 
addition the supematants were analysed for the ability to neutralise human TNFa in the 
receptor assay. 

6 lead clones (dl-d6 below) that neutralised in these assays and have been sequenced. 
The results shows that out of the 6 clones obtained there are only 3 different second dAbs 
(dAbl, dAb2 and dAb3) however where the second dAb is found more than once they are 
linked with different length linkers: 

TARl-5dl: 3U linker 2 nd dAb=dAbl - l|ug/ml Ag immunotube overnight wash 
TARl-5d2: 3U linker 2 nd dAb=dAb2 - l^g/ml Ag immunotube overnight wash 
TARl-5d3: 5U linker 2 nd dAb=dAb2 - lfig/ml Ag immunotube overnight wash 
TARl-5d4: 5U linker 2 nd dAb=dAb3 - 20(ag/ml Ag immunotube overnight wash 



WO 2004/003019 



PCT/GB2003/002804 



102 

TARl-5d5: 5U linker 2 nd dAb=dAbl - 20jJ,g/ml Ag immunotube overnight wash 
TARl-5d6: 7U linker 2 dAb=dAbl- Rl:l|ag/ml Ag immunotube overnight wash, 
R2: beads 



5 The 6 lead clones were examined further. Protein was produced from the periplasm and 
supernatant, purified with protein L agarose and examined in the cell and receptor assays. 
The levels of neutralisation were variable (Table 1). The optimal conditions for protein 
preparation were determined. Protein produced from HB2151 cells as supernatants gave 
the highest yield (approximately lOmgs/L of culture). The supernatants were incubated 
10 with protein L agarose for 2hrs at room temperature or overnight at 4°C. The beads were 
washed with PBS/NaCl and packed onto an FPLC column using a peristaltic pump. The 
beads were washed with 10 column volumes of PBS/NaCl and eluted with 0.1M glycine 
pH2.5. In general, dimeric protein is eluted after the monomer. 

15 TARl-5dl-6 dimers were purified by FPLC. Three species were obtained, by FPLC 
purification and were identified by SDS PAGE. One species corresponds to monomer and 
the other two species corresponds to dimers of different sizes. The larger of the two 
species is possibly due to the presence of C terminal tags. These proteins were examined 
in the receptor assay. The data presented in table 1 represents the optimum results 

20 obtained from the two dimeric species (Figure 1 1) 

The three second dAbs from the dimer pairs (ie, dAbl, dAb2 and dAb3) were cloned as 
monomers and examined by ELISA and in the cell and receptor assay. All three dAbs 
bind specifically to TNF by antigen ELISA and do not cross react with plastic or BSA. As 
25 monomers, none of the dAbs neutralise in the cell or receptor assays. 



2.1.2 TAR1-5-19 dimers 

TAR1-5-19 was substituted for TAR1-5 in the 6 lead clones. Analysis of all TAR1-5-19 
dimers in the cell and receptor assays was conducted using total protein (protein L 
30 purified only) unless otherwise stated (Table 2). TARl-5-19d4 and TARl-5-19d3 have 
the best ND50 (~5nM) in the cell assay, this is consistent with the receptor assay results 
and is an improvement over TAR1-5-19 monomer (ND 50 ~30nM). Although purified 
TAR1-5 dimers give variable results in the receptor and cell assays TAR1-5-19 dimers 



WO 2004/003019 



PCT/GB2003/002804 



103 

were more consistent. Variability was shown when using different elution buffers during 
the protein purification. Elution using 0.1M Phosphate-citrate buffer pH2.6 or 0.2M 
Glycine pH2.5 although removing all protein from the protein L agarose in most cases 
rendered it less functional. 

TARl-5-19d4 was expressed in the fermenter and purified on cation exchange FPLC to 
yield a completely pure dimer. As with TARl~5d4 three species were obtained, by FPLC 
purification corresponding to monomer and two dimer species. This dimer was amino 
acid sequenced. TAR 1-5- 19 monomer and TARl-5-19d4 were then examined in the 
receptor assay and the resulting IC50 for monomer was 30nM and for dimer was 8nM. 
The results of the receptor assay comparing TAR1-5-19 monomer, TARl-5-19d4 and 
TARl-5d4 is shown in figure 10. 

TAR1-5-19 homodimers were made in the 3U, 5U and 7U vectors, expressed and purified 
on Protein L. The proteins were examined in the cell and receptor assays and the resulting 
IC 5 os (for receptor assay) and ND 50 s (for cell assay) were determined (table 3, figure 12). 

2.2 Fabs 

TAR 1-5 and TAR1-5-19 dimers were also cloned into Fab format, expressed and purified 
on protein L agarose. Fabs were assessed in the receptor assays (Table 4). The results 
showed that for both TAR1-5-19 and TAR1-5 dimers the neutralisation levels were 
similar to the original Gly4Ser linker dimers from which they were derived. A TAR1-5-19 
Fab where TAR 1-5- 19 was displayed on both CH and CK was expressed, protein L 
purified and assessed in the receptor assay. The resulting IC50 was approximately InM. 

2.3 TAR1-27 dimers 

3 x 96 clones were picked from the round 2 selection encompassing all the libraries and 
selection conditions. 2ml supernatant preps were made for analysis in ELISA and 
bioassays. Antigen ELISA gave 71 positive clones. The receptor assay of crude 
supernatants yielded 42 clones with inhibitory properties (TNF binding 0-60%). In the 
majority of cases inhibitory properties correlated with a strong ELISA signal. 42 clones 



WO 2004/003019 PCT/GB2003/002804 

104 

were sequenced, 39 of these have unique second dAb sequences. The 12 dimers that 
gave the best inhibitory properties were analysed farther. 

The 12 neutralising clones were expressed as 200ml supernatant preps and purified on 
5 protein L. These were assessed by protein L and antigen ELISA, BIAcore and in the 
receptor assay. Strong positive ELISA signals were obtained in all cases. BIAcore 
analysis revealed all clones to have fast on and off rates. The off rates were improved 
compared to monomelic TAR1-27, however the off rate of TAR 1-27 dimers was faster 
(Koff is approximately in the range of 10" 1 and 10* 2 M) than the TAR1-5 dimers examined 
10 previously (Koff is approximately in the range of 10' 3 - 10"* 4 M). The stability of the 
purified dimers was questioned and therefore in order to improve stability, the addition on 
5%glycerol, 0.5% Triton XI 00 or 0.5% NP40 (Sigma) was included in the purification of 

2 TAR1-27 dimers (d2 and dl6). Addition of NP40 or Triton X100™ improved the yield 
of purified product approximately 2 fold. Both dimers were assessed in the receptor 

15 assay. TARl-27d2 gave IC50 of ~30nM under all purification conditions. TARl-27dl6 
showed no neutralisation effect when purified without the use of stabilising agents but 
gave an IC50 of ~50nM when purified under stabilising conditions. No further analysis 
was conducted. 

20 2.4 TAR2-5 dimers 

3 x 96 clones were picked from the second round selections encompassing all the libraries 
and selection conditions. 2ml supernatant preps were made for analysis. Protein A and 
antigen ELISAs were conducted for each plate. 30 interesting clones were identified as 
having good off-rates by BIAcore (Koff ranges between 10* 2 - 10" 3 M). The clones were 

25 sequenced and 13 unique dimers were identified by sequence analysis. 



Table 1: TAR1-5 dimers 



Dimer 


Cell 
type 


Purification 


Protein 
Fraction 


Elution 
conditions 


Receptor/ 
Cell assay 


TARl-5dl 


HB2151 


Protein L + 
FPLC 


small dimeric 
species 


0.1M glycine 
pH2.5 


RA~30nM 


TARl-5d2 


HB2151 


Protein L + 
FPLC 


small dimeric 
species 


0.1M glycine 
pH2.5 


RA~50nM 



WO 2004/003019 



105 



PCT/GB2003/002804 







FPLC 


species 


pH2.5 

EL 


M 


TARl-5d3 


HB2151 


Protein L + 
FPLC 


large dimeric 
species 


0.1M glycine 
pH2.5 


RA-300 
nM 


TARl-5d4 


HB2151 


Protein L + 
FPLC 


small dimeric 
species 


0 . 1 M glycine 
pH2.5 


RA~3n 
M 


TARl-5d5 


HB2151 


Protein L + 
FPLC 


large dimeric 
species 


0.1M glycine 
pH2.5 


RA-200 
nM 


TARl-5d6 


HB2151 


Protein L 
+FPLC 


Large dimeric 
species 


0.1M glycine 
pH2.5 


RA-100 
nM 



'•'note dimer 2 and dimer 3 have the same second dAb (called dAb2), however have 
different linker lengths (d2 = (Gly 4 Ser) 3 , d3 = (Gly 4 Ser) 3 ). dAbl is the partner dAb to 
dimers 1, 5 and 6. dAb3 is the partner dAb to dimer4. None of the partner dAbs 
5 neutralise alone. FPLC purification is by cation exchange unless otherwise stated. The 

optimal dimeric species for each dimer obtained by FPLC was determined in these 
assays. 



10 Table 2: TAR1-5-19 dimers 



Dimer 


Cell type 


Purification 


Protein 
Fraction 


Eiution conditions 


Recept 
or/ Cell 

assay 


TAR1-5-19 dl 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH 2.0 


RA-15 
nM 


TAR1-5-19 d2 (no 
stop codon) 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH 2.0 + 
0.05%NP40 


RA~2n 
M 


TARl-5-19d3 
(no stop codon) 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH 2.5.+ 
0.05%NP40 


RA~8n 
M 


TARl-5-19d4 


TOPI OF' 


Protein L + 
FPLC 


FPLC purified 
fraction 


0.1M glycine 
pH2.0 


RA~2- 
5nM 
CA-12 
nM 


TARl-5-19d5 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH2.0 + 
NP40 


RA~8n . 
M 

CA-10 
nM 


TAR1-5-19 d6 


TOPI OF' 


Protein L 


Total protein 


0.1M glycine pH2.0 

* 


RA-10 
nM 



Table 3: TAR1-5-19 homodimers 

15 



Dimer 


Cell type 


Purification 


Protein Fraction 


Eiution conditions 


Recept 








or/ Cell 












assay 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



106 



homodimer 








pH2.5 


CA- 


-3nM 


TAR1-5-19 7U 
homodimer 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.5 


glycine 


RA- 
CAS 


40nM 
45nM 


TAR1-5-19 cys 
hinge 


HB2151 


Protein L + 
FPLC 


FPLC purified 
dimer fraction 


0.1M 
pH2.5 


glycine 


RA- 


-2nM 


TAR1-5- 
19CH/ TAR1- 
5-19 CK 


HB2151 


Protein 


Total protein 


0.1M 
pH2.5 


glycine 


RA- 


TnM 


Table 4: 


TAR1-5/TAR1-5-19 Fabs 












Dimer 


Cell 
type 


Purification 


Protein 
Fraction 


Elution 
conditions 


Receptor/ Cell 
assay 


TAR1-5CH/ 

JAM /^TZ" 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.6 


citrate 


RA 


~90nM 


TAR1-5CH/ 
dAb2 CK 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.5 


glycine 


RA 

CA- 


~30nM 
~60nM 


dAb3CH/ 
TAR1-5CK 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.6 


citrate 


RA- 


~100nM 


TAR1-5- 
19CH/ 
dAbl CK 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.0 


glycine 


RA 


-6nM 


dAbl CH/ 
TAR1-5-19CK 


HB2151 


Protein L 


0.1M glycine 
pH2.0 


Myc/flag 


RA- 


-6nM 


TAR1-5- 
19CH/ 
dAb2 CK 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.0 


glycine 


RA- 
CA- 


-8nM 
42nM 


TAR1-5- 

19CH/ 

dAb3CK 


HB2151 


Protein L 


Total protein 


0.1M 
pH2.0 


glycine 


RA- 


-3nM 



5 

Example 7 

dAb dimerisation by terminal cysteine linkage 
10 Summary 

For dAb dimerisation, a free cysteine has been engineered at the C-terminus of the 
protein. When expressed the protein forms a dimer which can be purified by a two step 
purification method. 



WO 2004/003019 



PCT/GB2003/002804 



107 

PCR construction of TAR1-5-19CYS dimer 

See example 8 describing the dAb trimer. The trimer protocol gives rise to a mixture of 
monomer, dimer and trimer. 

5 Expression and purification of TAR1-5-19CYS dimer 

The dimer was purified from the supernatant of the culture by capture on Protein L 
agarose as outlined in the example 8. 

Separation of TAR1-5-19CYS monomer from the TAR1-5-19CYS dimer 

10 Prior to cation exchange separation, the mixed monomer/dimer sample was buffer 
exchanged into 50 mM sodium acetate buffer pH 4.0 using a PD-10 column (Amersham 
Pharmacia), following the manufacturer's guidelines. The sample was then applied to a 
lmL Resource S cation exchange column (Amersham Pharmacia), which had been pre- 
equilibrated with 50 mM sodium acetate pH 4.0. The monomer and dimer were separated 

15 using the following salt gradient in 50 mM sodium acetate pH 4.0: 

150 to 200 mM sodium chloride over 15 column volumes 
200 to 450 mM sodium chloride over 10 column volumes 
450 to 1000 mM sodium chloride over 15 column volumes 

20 

Fractions containing dimer only were identified using SDS-PAGE and then pooled and 
. the pH increased to 8 by the addition of 1/5 volume of 1M Tris pH 8.0. 

In vitro functional binding assay: TNF receptor assay and cell assay 
25 The affinity of the dimer for human TNFqj was determined using the TNF receptor and 
cell assay. IC50 in the receptor assay was approximately 0.3-0.8 nM; ND50 in the cell 
assay was approximately 3-8 nM. 

Other possible TAR1-5-19CYS dimer formats 

30 

PEG dimers and custom synthetic maleimide dimers 

Nektar (Shearwater) offer a range of bi-maleimide PEGs [mPEG2-(MAL)2 or mPEG- 
(MAL)2] which would allow the monomer to be formatted as a dimer, with a small linker 



WO 2004/003019 



PCT/GB2003/002804 



108 

separating the dAbs and both being linked to a PEG ranging in size from 5 to 40 kDa. It 
has been shown that the 5kDa mPEG-(MAL)2 (ie, [TARl-5-19]-Cys-maleimide-PEG x 
2, wherein the maleimides are linked together in the dimer) has an affinity in the TNF 
receptor assay of - 1-3 nM. Also the dimer can also be produced using TMEA (Tris[2~ 
maleimidoethyl]amine) (Pierce Biotechnology) or other bi-functional linkers. 

It is also possible to produce the disulphide dimer using a chemical coupling procedure 
using 2,2'-dithiodipyridine (Sigma Aldrich) and the reduced monomer. 

Addition of a polypeptide linker or hinge to the C-terminus of the dAb. 
A small linker, either (Gly 4 Ser) n where n=l to 10, eg, 1, 2, 3, 4, 5, 6 or 7, an 
immunoglobulin (eg, IgG hinge region or random peptide sequence (eg, selected from a 
library of random peptide sequences) can be engineered between the dAb and the terminal 
cysteine residue. This can then be used to make dimers as outlined above. 

Example 8 

dAb trimerisation 

Summary 

For dAb trimerisation, a free cysteine is required at the C-terminus of the protein. The 
cysteine residue, once reduced to give the free thiol, can then be used to specifically 
couple the protein to a trimeric maleimide molecule, for example TMEA (Tris[2- 
maleimido ethyl] amine) . 

PGR construction of TAR1-5-19CYS 

The following oligonucleotides were used to specifically PGR TAR1-5-19 with a Sail and 
BarnHI sites for cloning and also to introduce a C-terminal cysteine residue: 

Sail 

Trp Ser Ala Ser Thr Asp* lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 
1 TGG AGC GCG TCG ACG GAC ATC CAG ATG ACC CAG TCT CCA TCC TCT CTG TCT GCA TCT GTA 
ACC TCG CGC AGC TGC CTG TAG GTC TAC TGG GTC AGA GGT AGG AGA GAC AGA CGT AGA CAT 



Gly Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Ser lie Asp Ser Tyr Leu His Trp 



WO 2004/003019 



PCT/GB2003/002804 



109 

61 GGA GAC CGT GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC ATT GAT AGT TAT TTA CAT TGG 
CCT CTG GCA CAG TGG TAG TGA ACG GCC CGT TCA GTC TCG TAA CTA TCA ATA AAT GTA ACC 





Tyr 


Gin 


Gin 


Lys 


Pro 


Gly 


Lys 


Ala 


Pro 


Lys 


Leu 


Leu 


lie 


Tyr 


Ser 


Ala 


Ser 


Glu 


Leu 


Gin 


121 


TAC 


CAG 


CAG 


AAA 


CCA 


GGG 


AAA 


GCC 


CCT 


AAG 


CTC 


CTG 


ATC 


TAT 


AGT 


GCA 


TCC 


GAG 


TTG 


CAA 




ft TVS 


V7 J. V_ 




XXX 


^3\J X 






err* 




X X 






T>7\ f* 
J.Ab 


Al A 


TCA 


CGT 


AGG 


CTC 


AAC 


GTT 




Ser 


Gly 


Val 


Pro 


Ser 


Arg 


Phe 


Ser 


Gly 


Ser 


Gly 


Ser 


Gly 


Thr 


Asp 


Phe 


Thr 


Leu 


Thr 


He 


181 


AGT 


GGG 


GTC 


CCA 


TCA 


CGT 


TTC 


AGT 


GGC 


AGT 


GGA 


TCT 


GGG 


ACA 


GAT 


TTC 


ACT 


CTC 


ACC 


ATC 




TCA 


CCC 


CAG 


GGT 


AGT 


GCA 


AAG 


TCA 


CCG 


TCA 


CCT 


AGA 


CCC 


TGT 


CTA 


AAG 


TGA 


GAG 


TGG 


TAG 




Ser 


Ser 


Leu 


Gin 


Pro 


Glu 


Asp 


Phe 


Ala 


Thr 


Tyr 


Tyr 


Cys 


Gin 


Gin 


Val 


Val 


Trp 


Arg 


Pro 


241 


AGC 


AGT 


CTG 


CAA 


CCT 


GAA 


GAT 


TTT 


GCT 


ACG 


TAC 


TAC 


TGT 


CAA 


CAG 


GTT 


GTG 


TGG 


CGT 


CCT 




TCG 


TCA 


GAC 


GTT 


GGA 


CTT 


CTA 


AAA 


CGA 


TGC 


ATG 


ATG 


ACA 


GTT 


GTC 


CAA 


CAC 


ACC 


GCA 


GGA 




































BamHI 








Phe 


Thr 


Phe 


Gly 


Gin 


Gly 


Thr 


Lys 


Val 


Glu 


lie 


Lys 


Arg 


Cys 


*** 


* ** 


~ ~ -w M _ 

Gly Ser 


Gly 




301 


TTT 


ACG 


TTC 


GGC 


CAA 


GGG 


ACC 


AAG 


GTG 


GAA 


ATC 


AAA 


CGG 


TGC 


TAA 


TAA 


GGA 


TCC 


GGC 






AAA 


TGC 


AAG 


CCG 


GTT 


CCC 


TGG 


TTC 


CAC 


CTT 


TAG 


TTT 


GCC 


ACG 


ATT 


ATT 


CCT 


AGG 


CCG 





(* start of TAR1-5-19CYS sequence) 
Forward primer 

5 ' -TGGAGCGCGTC GAC GGAC ATC C AGATGAC C C AGTCTC C A-3 ' 
Reverse primer 

5 '-TTAGCAGCCGGATCCTTATTAGCACCGTTTGATTTCCAC-3 * 

The PCR reaction (50\xL volume) was set up as follows: 200jliM dNTPs, 0.4jnM of each 
primer, 5 jliL of lOx P/wTurbo buffer (Stratagene), 100 ng of template plasmid (encoding 
TAR 1-5- 19), l\xL of PfuTvxbo enzyme (Stratagene) and the volume adjusted to 50|liL 
using sterile water. The following PCR conditions were used: initial denaturing step 94 
°C for 2 mins, then 25 cycles of 94 °C for 30 sees, 64 °C for 30 sec and 72 °C for 30 sec. 
A final extension step was also included of 72 °C for 5 mins. The PCR product was 
purified and digested with Sail and BamHI and ligated into the vector which had also 
been cut with the same restriction enzymes. Correct clones were verified by DNA 
sequencing. 



WO 2004/003019 PCT/GB2003/002804 

110 

Expression and purification of TAR1-5-19CYS 

TAR1-5-19CYS vector was transformed into BL21 (DE3) pLysS chemically competent 
cells (Novagen) following the manufacturer's protocol. Cells carrying the dAb plasmid 
were selected for using lOOpg/mL carbenicillin and 37 jig/mL chloramphenicol. Cultures 

5 were set up in 2L baffled flasks containing 500 mL of terrific broth (Sigma- Aldrich), 
lOOpg/mL carbenicillin and 37 pg/mL chloramphenicol. The cultures were grown at 30 
°C at 200rpm to an O.D.600 of 1-1.5 and then induced with ImM EPTG (isopropyl-beta- 
D~thiogalactopyranoside, from Melford Laboratories). The expression of the dAb was 
allowed to continue for 12-16 hrs at 30 °C. It was found that most of the dAb was present 

10 in the culture media. Therefore, the cells were separated from the media by centrifagation 
(8,000xg- for 30 mins), and the supernatant used to purify the dAb. Per litre of 
supernatant, 30 mL of Protein L agarose (Affitech) was added and the dAb allowed to 
batch bind with stirring for 2 hours. The resin was then allowed to settle under gravity for 
a further hour before the supernatant was siphoned off The agarose was then packed into 

15 a XK 50 column (Amersham Phamacia) and was washed with 10 column volumes of 
PBS. The bound dAb was eluted with 100 mM glycine pH 2.0 and protein containing 
fractions were then neutralized by the addition of 1/5 volume of 1 M Tris pH 8.0. Per litre 
of culture supernatant 20 mg of pure protein was isolated, which contained a 50:50 ratio 
of monomer to dimer. 

20 

Trimerisation of TAR1-5-19CYS 

2.5 ml of 100 pM TAR1-5-19CYS was reduce with 5 mM dithiothreitol and left at room 
temperature for 20 minutes. The sample was then buffer exchanged using a PD-10 
column (Amersham Pharmacia). The column had been pre-equilibrated with 5 mM 

25 EDTA, 50 mM sodium phosphate pH 6.5, and the sample applied and eluted following 

the manufactures guidelines. The sample was placed on ice until required. TMEA (Tris[2- 
maleimidoethyl] amine) was purchased from Pierce Biotechnology. A 20 mM stock 
solution of TMEA was made in 100% DMSO (dimethyl sulphoxide). It was found that a 
concentration of TMEA greater than 3 : 1 (molar ratio of dAb:TMEA) caused the rapid 

30 precipitation and cross-linking of the protein. Also the rate of precipitation and cross- 
linking was greater as the pH increased. Therefore using 100 pM reduced TAR1-5- 
19CYS, 25 pM TMEA was added to trimerise the protein and the reaction allowed to 
proceed at room temperature for two hours. It was found that the addition of additives 



WO 2004/003019 



PCT/GB2003/002804 



111 

such as glycerol or ethylene glycol to 20% (v/v), significantly reduced the precipitation 
of the trimer as the coupling reaction proceeded. After coupling, SDS-PAGE analysis 
showed the presence of monomer, dimer and trimer in solution. 

Purification of the trimeric TAR1-5-19CYS 

40 |J,L of 40% glacial acetic acid was added per mL of the TMEA-TARl-5-19cys reaction 
to reduce the pH to ~4. The sample was then applied to a lmL Resource S cation 
exchange column (Amersham Pharmacia), which had been pre-equilibrated with 50 mM 
sodium acetate pH 4.0. The dimer and trimer were partially separated using a salt gradient 
of 340 to 450 mM Sodium chloride, 50 mM sodium acetate pH 4.0 over 30 column 
volumes. Fractions containing trimer only were identified using SDS-PAGE and then 
pooled and the pH increased to 8 by the addition of 1/5 volume of 1M Tris pH 8.0. To 
prevent precipitation of the trimer during concentration steps (using 5K cut off Viva spin 
concentrators; Vivascience), 10% glycerol was added to the sample. 

In vitro functional binding assay: TNF receptor assay and cell assay 
The affinity of the trimer for human TNFa was determined using the TNF receptor and 
cell assay. IC50 in the receptor assay was 0.3nM; ND50 in the cell assay was in the range 
of 3 to lOnM (eg, 3nM). 

Other possible TAR1-5-19CYS trimer formats 

TAR1-5-19CYS may also be formatted into a trimer using the following reagents: 
PEG trimers and custom synthetic maleimide trimers 

Nektar (Shearwater) offer a range of multi arm PEGs, which can be chemically modified 
at the terminal end of the PEG. Therefore using a PEG trimer with a maleimide functional 
group at the end of each arm would allow the trimerisation of the dAb in a manner similar 
to that outlined above using TMEA. The PEG may also have the advantage in increasing 
the solubility of the trimer thus preventing the problem of aggregation. Thus, one could 
produce a dAb trimer in which each dAb has a C-terminal cysteine that is linked to a 
maleimide functional group, the maleimide functional groups being linked to a PEG 
trimer. 



WO 2004/003019 



PCT/GB2003/002804 



112 

Addition of a polypeptide linker or hinge to the C-terminus of the dAb 
A small linker, either (Gly 4 Ser) n where n= 1 to 10, eg, 1, 2, 3, 4, 5, 6 or 7 , an 
immunoglobulin (eg, IgG hinge region or random peptide sequence (eg, selected from a 
library of random peptide sequences) could be engineered between the dAb and the 
5 terminal cysteine residue. When used to make multimers (eg, dimers or trimers), this 
again would introduce a greater degree of flexibility and distance between the individual 
monomers, which may improve the binding characteristics to the target, eg a multisubunit 
target such as human TNFo;. 

10 Example 9. 

Selection of a collection of single domain antibodies (dAbs) directed against human 
serum albumin (HSA) and mouse serum albumin (MSA). 

15 This example explains a method for making a single domain antibody (dAb) directed 
against serum albumin. Selection of dAbs against both mouse serum albumin (MSA) and 
human serum albumin (HSA) is described. Three human phage display antibody libraries 
were used in this experiment, each based on a single human framework for V H (see 
Figure 13: sequence of dummy V H based on V3-23/DP47 and JH4b) or Vk (see Figure 

20 15: sequence of dummy Vk based on ol2/o2/DPK9 and Jkl) with side chain diversity 
encoded by NNK codons incorporated in complementarity determining regions (CDR1, 
CDR2 and CDR3). 

Library 1 (Vh): 

25 Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, 
H97, H98. 

Library size: 6.2 x 10 
Library 2 (V H ): 

30 Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, 
H97, H98, H99, H100, HlOOa, HlOOb. 
Library size: 4.3 x 10 



WO 2004/003019 



PCT/GB2003/002804 



113 

Library 3 (Vk): 

Diversity at positions: L30, L31, L32, L34, L50, L53, L91, L92, L93, L94, L96 
Library size: 2 x 1 0 9 

5 The V H and Vk libraries have been preselected for binding to generic ligands protein A 
and protein L respectively so that the majority of clones in the unselected libraries are 
functional. The sizes of the libraries shown above correspond to the sizes after 
preselection. 



10 Two rounds of selection were performed on serum albumin using each of the libraries 
separately. For each selection, antigen was coated on immunotube (nunc) in 4ml of PBS 
at a concentration of lOOjLCg/ml. hi the first round of selection, each of the three libraries 
was panned separately against HSA (Sigma) and MSA (Sigma). In the second round of 
selection, phage from each of the six first round selections was panned against (i) the 

15 same antigen again (eg 1 st round MSA, 2 nd round MSA) and (ii) against the reciprocal 
antigen (eg 1 st round MSA, 2 nd round HSA) resulting in a total of twelve 2 nd round 
selections. In each case, after the second round of selection 48 clones were tested for 
binding to HSA and MSA. Soluble dAb fragments were produced as described for scFv 
fragments by Harrison et al, Methods Enzymol. 1996;267:83-109 and standard ELISA 

20 protocol was followed (Hoogenboom et al (1991) Nucleic Acids Res., 19: 4133) except 
that 2% tween PBS was used as a blocking buffer and bound dAbs were detected with 
either protein L-HRP (Sigma) (for the Vks) and protein A -HRP (Amersham Pharmacia 
Biotech) (for the Vrs). 

25 dAbs that gave a signal above background indicating binding to MSA, HSA or both were 
tested in ELISA insoluble form for binding to plastic alone but all were specific for serum 
albumin. Clones were then sequenced (see table below) revealing that 21 unique dAb 
sequences had been identified. The minimum similarity (at the amino acid level) between 
the Vk dAb clones selected was 86.25% ((69/80)xl00; the result when all the diversified 

30 residues are different, eg clones 24 and 34). The minimum similarity between the V H dAb 
clones selected was 94 % ((127/136)xl00). 



WO 2004/003019 



PCT/GB2003/002804 



114 

Next, the serum albumin binding dAbs were tested for their ability to capture 
biotinylated antigen from solution. ELISA protocol (as above) was followed except that 
ELIS A plate was coated with 1 jug/ml protein L (for the V/c clones) and 1 /xg/ml protein A 
(for the V H clones). Soluble dAb was captured from solution as in the protocol and 
5 detection was with biotinylated MSA or HSA and streptavidin HRP. The biotinylated 
MSA and HSA had been prepared according to the manufacturer's instructions, with the 
aim of achieving an average of 2 biotins per serum albumin molecule. Twenty four 
clones were identified that captured biotinylated MSA from solution in the ELISA. Two 
of these (clones 2 and 38 below) also captured biotinylated HSA. Next, the dAbs were 
10 tested for their ability to bind MSA coated on a CMS biacore chip. Eight clones were 
found that bound MSA on the biacore. 



dAb fall 










XSIUGS 


PQTlf 11T*A 










A/TO A 
JVlbA 


biotinvlated 


H 








• 

in 


MSA1 


or k CDR1 






g~% f% /~\ 

Diacore : 


Vk library 3 












template 












(dummy) 


K 


XXXLX 


XASXLQS 


QQXXXXPXT 




2, 4, 7, 41, 


K 


SSYLN 


RASPLQS 


QQTYSVPPT 




38, 54 


K 


SSYLN 


RASPLQS 


QQTYRIPPT 




46, 47, 52, 56 


K 


FKSLK 


NASYLQS 


QQWYWPVT 




13, 15 


K 


YYHLK 


KASTLQS 


QQVRKVPRT 




30, 35 


K 


RRYLK 


QASVLQS 


QQGLYPPIT 




19, 


K 


YNWLK 


RASSLQS 


QQNWIPRT 




22, 


K 


LWHLR 


HASLLQS 


QQSAVYPKT 




23, 


K 


FRYLA 


HASHLQS 


QQRLLYPKT 




24, 


K 


FYHLA 


PASKLQS 


QQRARWPRT 




31, 


K 


IWHLN 


RASRLQS 


QQVARVPRT 




33, 


K 


YRYLR 


KASSLQS 


QQYVGYPRT 




34, 


K 


LKYLK 


NASHLQS 


QQTTYYPIT 




53, 


K 


LRYLR 


KASWLQS 


QQVLYYPQT 




11, 


K 


LRSLK 


AASRLQS 


QQWYWPAT 




12, 


K 


FRHLK 


AASRLQS 


QQVALYPKT 


v 


17, 


K 


RKYLR 


TASSLQS 


QQNLFWPRT 




18, 


K 


RRYLN 


AASSLQS 


QQMLFYPKT 


v 


16, 21 


K 


IKHLK 


GASRLQS 


QQGARWPQT 




25, 26 


K 


YYHLK 


KASTLQS 


QQVRKVPRT 




27, 


K 


YKHLK 


NASHLQS 


QQVGRYPKT 




55, 


K 


FKSLK 


NASYLQS 


QQWYWPVT 




V H library 1 












(and 2) 












template 












(dummy) 


H 


XXYXXX 


XIXXXGXXTXYADSVKG 


XXXX (XXXX) FDY 




8,10 


H 


WVYQMD 


S I S AFGAKTL YADS VKG 


LSGKFDY 




36, 


H 


WSYQMT 


SIS S FGS STLYADS VKG 


GRDHNYSLFDY 





Captures 
biotinylated 



S all 4 bind 
S both bind 



WO 2004/003019 



PCT/GB2003/002804 



115 

In all cases the frameworks were identical to the frameworks in the corresponding 
dummy sequence, with diversity in the CDRs as indicated in the table above. 

Of the eight clones that bound MSA on the biacore, two clones that are highly expressed 
5 in E. coli (clones MSA16 and MSA26) were chosen for further study (see example 10). 
Full nucleotide and amino acid sequences for MSA16 and 26 are given in figure 16. 

Example 10. 

10 Determination of affinity and serum half-life in mouse of MSA binding dAbs MSA16 
and MSA26. 

dAbs MSA16 and MSA26 were expressed in the periplasm of E. coli and purified using 
batch absorbtion to protein L-agarose affinity resin (Affitech, Norway) followed by 

15 elution with glycine at pH 2.2. The purified dAbs were then analysed by inhibition 
biacore to determine K<i. Briefly, purified MSA16 and MSA26 were tested to determine 
the concentration of dAb required to achieve 200RUs of response on a biacore CM5 chip 
coated with a high density of MSA. Once the required concentrations of dAb had been 
determined, MSA antigen at a range of concentrations around the expected Kd was 

20 premixed with the dAb and incubated overnight. Binding to the MSA coated biacore chip 
of dAb in each of the premixes was then measured at a high flow-rate of 30 jul/minute. 
The resulting curves were used to create Klotz plots, which gave an estimated Kid of 
200nM for MSA16 and 70nM for MSA 26 (Figure 17 A & B). 

25 Next, clones MSA16 and MSA26 were cloned into an expression vector with the HA tag 
(nucleic acid sequence: TATCCTTATGATGTTCCTGATTATGCA and amino acid 
sequence: YPYDVPDYA) and 2-10 mg quantities were expressed in E. coli and purified 
from the supernatant with protein L-agarose affinity resin (Affitech, Norway) and eluted 
with glycine at pH2.2. Serum half life of the dAbs was determined in mouse. MSA26 

30 and MSA16 were dosed as single i.v. injections at approx 1.5mg/kg into CD1 mice. 
Analysis of serum levels was by goat anti-HA (Abeam, UK) capture and protein L-HRP 
(invitrogen) detection ELISA which was blocked with 4% Marvel. Washing was with 
0.05% tween PBS. Standard curves of known concentrations of dAb were set up in the 



WO 2004/003019 



PCT/GB2003/002804 



116 

presence of Ixmouse serum to ensure comparability with the test samples. Modelling 
with a 2 compartment model showed MSA-26 had a tl/2ce of 0.1 6hr, a tl/2/3 of 14.5hr and 
an area under the curve (AUC) of 465hr.mg/ml (data not shown) and MSA- 16 had a tl/2a 
of 0.98hr, a tl/2/3 of 36.5hr and an AUC of 913hr.mg/ml (figure 18). Both anti-MSA 
5 clones had considerably lengthened half life compared with HEL4 (an anti-hen egg white 
lysozyme dAb) which had a tl/2a of 0.06hr, and a tl/2/3 of 0.34hr. 

Example 11. 

10 Creation of V H -V H and V7e- Vk dual specific Fab like fragments 

This example describes a method for making Vh- Vh and V/c-V/c dual specifics as Fab 
like fragments. Before constructing each of the Fab like fragments described, dAbs that 
bind to targets of choice were first selected from dAb libraries similar to those described 

15 in example 9. A V H dAb, HEL4, that binds to hen egg lysozyme (Sigma) was isolated 
and a second Vh dAb (TAR2h-5) that binds to TNFa receptor (R and D systems) was also 
isolated. The sequences of these are given in the sequence listing. A Vk dAb that binds 
TNFa (TAR1-5-19) was isolated by selection and affinity maturation and the sequence is 
also set forth in the sequence listing. A second Vk dAb (MSA 26) described in example 9 

20 whose sequence is in figure 17B was also used in these experiments. 

DNA from expression vectors containing the four dAbs described above was digested 
with enzymes Sail and NotI to excise the DNA coding for the dAb. A band of the 
expected size (300-400bp) was purified by running the digest on an agarose gel and 
25 excising the band, followed by gel purification using the Qiagen gel purification kit 
(Qiagen, UK). The DNA coding for the dAbs was then inserted into either the Ch or Ck 
vectors (Figs 8 and 9) as indicated in the table below. 



dAb 


Target antigen 


dAb V H or 
dAb Vk 


Inserted into 
vector 


tag(C 
terminal) 


Antibiotic 
resisitance 


HEL4 


Hen egg lysozyme 


V H 


C H 


myc 


Chloramphenicol j 


TAR2-5 


TNF receptor 


V h 


Ck 


flag 


Ampicillin 


TAR1-5-19 


TNF a 


Vk 


C h 


myc 


Chloramphenicol 


MSA 26 


Mouse serum albumin 


Vk 


Ck 


flag 


Ampicillin 



WO 2004/003019 



PCT/GB2003/002804 



117 

The V H C h and Vh Ck constructs were cotransformed into HB2151 cells. Separately, the 
V/c Ch and Vk Ck constructs were cotransformed into HB2151 cells. Cultures of each of 
the cotransformed cell lines were grown overnight (in 2xTy containing 5% glucose, 
10jUg/ml chloramphenicol and 100/ig/ml ampicillin to maintain antibiotic selection for 
5 both C H and Ck plasmids). The overnight cultures were used to inoculate fresh media 
(2xTy, 10/ig/ml chloramphenicol and 100jU,g/ml ampicillin) and grown to OD 0.7-0.9 
before induction by the addition of IPTG to express their C H and Ck constructs. 
Expressed Fab like fragment was then purified from the periplasm by protein A 
purification (for the contransformed V H C H and V H Ck) and MSA affinity resin 
10 purification (for the contransformed V/c Ch and Vk Ck). 



Vh-Vh dual specific 

Expression of the V H Ch and V H Ck dual specific was tested by running the protein on a 
gel. The gel was blotted and a band the expected size for the Fab fragment could be 

15 detected on the Western blot via both the myc tag and the flag tag, indicating that both the 
V H C h and V H Ck parts of the Fab like fragment were present. Next, in order to 
determine whether the two halves of the dual specific were present in the same Fab-like 
fragment, an ELISA plate was coated overnight at 4°C with 100 /jlI per well of hen egg 
lysozyme (HEL) at 3 mg/ml in sodium bicarbonate buffer. The plate was then blocked 

20 (as described in example 1) with 2% tween PBS followed by incubation with the V H Ch 
/V h Ck dual specific Fab like fragment. Detection of binding of the dual specific to the 
HEL was via the non cognate chain using 9el0 (a monoclonal antibody that binds the 
myc tag, Roche) and anti mouse IgG-HRP (Amersham Pharmacia Biotech). The signal 
for the V H C h /V h Ck dual specific Fab like fragment was 0.154 compared to a 

25 background signal of 0.069 for the V H Ck chain expressed alone. This demonstrates that 
the Fab like fragment has binding specificity for target antigen. 



V K -V K dual specific 

After purifying the contransformed V/c C H and Vk Ck dual specific Fab like fragment on 
30 an MSA affinity resin, the resulting protein was used to probe an ELISA plate coated with 
lptg/ml TNFa and an ELISA plate coated with lO/xg/ml MSA. As predicted, there was 
signal above background when detected with protein L-HRP on bot ELISA plates (data 
not shown). This indicated that the fraction of protein able to bind to MSA (and therefore 



WO 2004/003019 



PCT/GB2003/002804 



118 

purified on the MSA affinity column) was also able to bind TNFa in a subsequent 
ELISA, confirming the dual specificity of the antibody fragment. This fraction of protein 
was then used for two subsequent experiments. Firstly, an ELISA plate coated with 
, 1/^g/ml TNFa was probed with dual specific V/c Ch and V/c C/c Fab like fragment and 
also with a control TNFa binding dAb at a concentration calculated to give a similar 
signal on the ELISA. Both the dual specific and control dAb were used to probe the 
ELISA plate in the presence and in the absence of 2mg/ml MSA. The signal in the dual 
specific well was reduced by more than 50% but the signal in the dAb well was not 
reduced at all (see figure 19a). The same protein was also put into the receptor assay with 
and without MSA and competition by MSA was also shown (see figure 19c). This 
demonstrates that binding of MSA to the dual specific is competitive with binding to 
TNFa. 

Example 12. 

Creation of a Vk- Vk dual specific cys bonded dual specific with specificity for 
mouse serum albumin and TNFa 

This example describes a method for making a dual specific antibody fragment specific 
for both mouse serum albumin and TNFa by chemical coupling via a disulphide bond. 
Both MSA16 (from example 1) and TAR1-5-19 dAbs were recloned into a pET based 
vector with a C terminal cysteine and no tags. The two dAbs were expressed at 4-10 mg 
levels and purified from the supernatant using protein L-agarose affinity resin (Affitiech, 
Norway). The cysteine tagged dAbs were then reduced with dithiothreitol. The TAR1-5- 
1 9 dAb was then coupled with dithiodipyridine to block reformation of disulphide bonds 
resulting in the formation of PEP 1-5-19 homodimers. The two different dAbs were then 
mixed at pH 6.5 to promote disulphide bond formation and the generation of TAR1-5-19, 
MSA16 cys bonded heterodimers. This method for producing conjugates of two unlike 
proteins was originally described by King et al (King TP, Li Y Kochoumian L 
Biochemistry. 1978 vol 17: 1499-506 Preparation of protein conjugates via intermolecular 
disulfide bond formation.) Heterodimers were separated from monomeric species by 
cation exchange. Separation was confirmed by the presence of a band of the expected 



WO 2004/003019 



PCT/GB2003/002804 



119 

size on a SDS geL The resulting heterodimeric species was tested in the TNF receptor 
assay and found to have an IC50 for neutralising TNF of approximately 1 8 nM. Next, 
the receptor assay was repeated with a constant concentration of heterodimer (18nM) and 
a dilution series of MSA and HSA. The presence of HSA at a range of concentrations (up 
5 to 2 mg/ml) did not cause a reduction in the ability of the dimer to inhibit TNFa . 
However, the addition of MSA caused a dose dependant reduction in the ability of the 
dimer to inhibit TNFa (figure 20).This demonstrates that MSA and TNFa compete for 
binding to the cys bonded TAR1-5-19, MSA16 dimer. 

10 Data Summary 

A summary of data obtained in the experiments set forth in the foregoing examples is set 
forth in Annex 4. 

All publications mentioned in the present specification, and references cited in said 
15 publications, are herein incorporated by reference. Various modifications and variations 
of the described methods and system of the invention will be apparent to those skilled in 
the art without departing from the scope and spirit of the invention. Although the 
invention has been described in connection with specific preferred embodiments, it 
should be understood that the invention as claimed should not be unduly limited to such 
20 specific embodiments. Indeed, various modifications of the described modes for carrying 
out the invention which are obvious to those skilled in molecular biology or related fields 
are intended to be within the scope of the following claims. 



WO 2004/003019 



PCT/GB2003/002804 



120 

Annex 1; polypeptides which enhance half-life in vivo. 

Alpha- 1 Glycoprotein (Orosomucoid) (AAG) 

Alpha- 1 Antichyromotrypsin (ACT) 

Alpha- 1 Antitrypsin (AAT) 

Alpha- 1 Microglobulin (Protein HC) (AIM) 

Alpha-2 Macroglobulin (A2M) 

Antithrombin III (AT III) 

Apolipoprotein A-l (Apo A-l) 

Apoliprotein B (Apo B) 

Beta-2-microglobulin (B2M) 

Ceruloplasmin (Cp) 

Complement Component (C3) 

Complement Component (C4) 

CI Esterase Inhibitor (CI INH) 

C-Reactive Protein (CRP) 

Cystatin C (Cys C) 

Ferritin (FER) 

Fibrinogen (FIB) 

Fibronectin (FN) 

Haptoglobin (Hp) 

Hemopexin (HPX) 

Immunoglobulin A (IgA) 

Immunoglobulin D (IgD) 

Immunoglobulin E (IgE) 

Immunoglobulin G (IgG) 

Immunoglobulin M (IgM) 

Immunoglobulin Light Chains (kapa/lambda) 

Lipoprotein(a) [Lp(a)] 

Mannose-bindign protein (MBP) 

Myoglobin (Myo) 

Plasminogen (PSM) 

Prealbumin (Transthyretin) (PAL) 



WO 2004/003019 



PCT/GB2003/002804 



121 

Retinol-binding protein (RBP) 
Rheomatoid Factor (RF) 
Serum Amyloid A (SAA) 
Soluble Tranferrin Receptor (sTfR) 
5 Transferrin (Tf) 



WO 2004/003019 



PCT/GB2003/002804 



122 

Annex 2 



Pairing 


Therapeutic relevant references. 


TNF 

ALPHA/TGF-P 


• TGF-b and TNF when injected into the ankle joint of collagen induced 
arthritis model significantly enhanced joint inflammation. In non-collagen 
challenged mice there was no effect. 


TNF ALPHA/IL- 
1 


• TNF and IL-1 synergize in the pathology of uveitis. 

• TNF and IL-1 synergize in the pathology of malaria (hypoglycaemia, NO). 

• TNF and IL-1 synergize in the induction of polymorphonuclear (PMN) 
cells migration in inflammation. 

• IL-1 and TNF synergize to induce PMN infiltration into the peritoneum. 

• IL-1 and TNF synergize to induce the secretion of IL-1 by endothelial cells. 
Important in inflammation. 

• IL-1 or TNF alone induced some cellular infiltration into knee synovium. 
IL-1 induced PMNs, TNF — monocytes, together tney maucea a more 
severe infiltration due to increased PMNs. 

• Circulating myocardial depressant substance (present in sepsis) is low 
levels of IL-1 and TNFacting synergistically. 


TNF ALPHA/IL-2 


• Most relating to synergistic activation of killer T-cells. 


TNF ALPHA/IL-3 


• Synergy of interleukin 3 and tumor necrosis factor alpha in stimulating 
clonal growth of acute myelogenous leukemia blasts is the result or 
induction of secondary hematopoietic cytokines by tumor necrosis factor 
alpha. 

• Cancer Res. 1992 Apr 15;52(8):2197-201. 


TNF ALPHA/IL-4 


• IL-4 and TNF synergize to induce VCAM expression on endothelial cells. 
Implied to have a role in asthma. Same for synovium - implicated in RA. 

• TNF and IL-4 synergize to mduce 1L-6 expression m Keratrnocytes. 

• Sustained elevated levels of VCAM- 1 in cultured fibroblast-like 
synoviocytes can be achieved by TNF-alpha in combination with either IL- 
4 or IL-13 through increased rriRNA stability. Am J Pathol. 1999 
Apr;154(4):l 149-58 


TNF ALPHA/IL-5 


• Relationship between the tumor necrosis factor system and the serum 

mterleukm-4, mterleuKin-3, inter leuKm- o , eosrnopnii canonic protein, ana 
immunoglobulin Jb levels in tne uroncniai nypcrreaciiviLy ui duun- oiiu 
their children. Allergy Asthma Proc. 2003 Mar-Apr ;24(2): 11 1-8. 


rr-vv T"|-» A T* T>TT A /TT /T 

TNF ALPHA/IL-o 


• 1JN-T and. lJL»-o are potent growtn iactors ior un-z, <x uuvci xiu.iiia.ii mycioma 
cell line. Eur J Haematol 1994 Jul;53(l):3 1-7. 


TNF ALPHA/IL-8 


• TNF and IL-8 synergized with PMNs to activate platelets. Implicated in 
Acute Respiratory .Distress oynarome. 

• See IL-5/TNF (asthma). Synergism between interleukin-8 and tumor 
necrosis factor-alnha for neutrophil-mediated platelet activation. Eur 
Cytokine Netw. 1994 Sep-Oct;5(5):455-60. (adult respiratory distress 
syndrome (ARDS)) 


TNF ALPHA/IL-9 




TNF ALPHA/IL- 
10 


• IL-10 induces and synergizes with TNF in the induction of HIV expression 
in chronically infected T-cells. 


TNF ALPHA/IL- 
11 


• Cytokines synergistically induce osteoclast differentiation: support by 
immortalized or normal calvarial cells. Am J Physiol Cell Physiol 2002 
Sep;283(3):C679-87. (Bone loss) 


TNF ALPHA/IL- 
12 





WO 2004/003019 



PCT/GB2003/002804 



123 



TNF ALPHA/IL- 
13 


• Sustained elevated levels of VCAM-1 in cultured fibroblast-like 
synoviocytes can be achieved by TNF-alpha in combination with either IL- 
4 or IL-13 through increased miviN A. staoiiity. Am J Jratnoi. 

Apr; 154(4): 1149-58. 

• Interleulcin- 1 i and tumour necrosis iactor-arpna synergisuoaiiy lnouce 
eotaxin production in human nasal fibroblasts. Clin Exp Allergy. 2000 
iviar,ou^j jj'to-jj. 

• Interleukin-13 and tumour necrosis factor-alpha synergistically induce 
eotaxin production in human nasal fibroblasts. Clin Exp Allergy. 2000 
Mar;30(3):348-55 (allergic inflammation) 

• Implications of serum TNF-beta and IL-13 in the treatment response of 
childhood nephrotic syndrome. Cytokine. 2003 Feb 7;21(3): 155-9. 


TNF ALPHA/IL- 
14 


• Effects of inhaled tumour necrosis factor alpha in subjects with mild 
asthma. Thorax. 2002 Sep;57(9):774-8. 


TNF ALPHA/IL- 
15 


• Effects of inhaled tumour necrosis factor alpha m subjects witn mild 
asthma. Thorax. 2002 Sep;57(9):774-8. 


TNF ALPHA/IL- 
16 


• Tumor necrosis factor- alpha-induced synthesis of interleukin-16 in airway 
epithelial cells: priming for serotonin stimulation. Am J Respir Cell Mo I 
Biol. 2003 Mar;28(3):354-62. (airway inflammation) 

• Correlation of circulating interleukin 16 with proinflammatory cytokines in 
patients with rheumatoid arthritis. Rheumatology (Oxford). 2001 
Apr;40(4):474-5. No abstract available. 

• Interleukin 1 6 is up-regulated in Crohn's disease and participates in TNBS 
colitis in mice. Gastroenterology. 2000 Oct;119(4):972-82. 


TNF ALPHA/IL- 
17 


• Inhibition of interleukin- 17 prevents the development of arthritis in 
vaccinated mice challenged with Borrelia burgdorferi. Infect Immun. 2003 
Jun;71(6):3437-42. 

• Interleukin 17 synergises with tumour necrosis factor alpha to induce 
cartilage destruction in vitro. Ann Rheum Dis. 2002 Oct;61(10):870-6. 

• A role of GM-CSF in the accumulation of neutrophils in the airways caused 
by IL-17 and TNF-alpha. Eur Respir J. 2003 Mar;21(3):387-93. (Airway 
inflammation) 

• Abstract Interleukin- 1, tumor necrosis factor alpha, and interleukin- 17 
synergistically up-regulate mtnc oxide and prostaglandin E2 production m 
explants of human osteoarthritic knee menisci. Arthritis Rheum. 2001 
Sep;44(9):2078-83. 


TNF ALPHA/IL- 
18 


• Association of mterleukm-18 expression with enhanced levels or botti 
interleukin-lbeta and tumor necrosis factor alpha in knee synovial tissue of 
patients witn rneumatoia artnritis. /Lrinriiis Kneum. zuuj reD } **o^zj.jji/- 
47. 

• Abstract Elevated levels of interleukin- 1 8 and tumor necrosis factor-alpha 
m serum 01 patients witn type z Qiaoetes meintus. reiationsnip wiui aiaueuc 
nephropathy. Metabolism. 2003 May;52(5): 605-8. 


TNF ALPHA/IL- 
19 


• Abstract IL-19 induces production of IL-6 and TNF-alpha and results in 
cell apoptosis through TNF-alpha. J Immunol. 2002 Oct 15;169(8):4288- 
97. 


TNF ALPHA/IL- 
20 


• Abstract Cytokines: IL-20 - a new effector in skin inflammation. Curr Biol. 
2001 Jul 10;11(13):R53 1-4 


TNF 

ALPHA/ Complem 
ent 


• Inflammation and coagulation: implications for the septic patient. Clin 
Infect Dis. 2003 May 15;36(10): 1259-65. Epub 2003 May 08. Review. 


TNF 

ALPHA/IFN-y 


• MHC induction in the brain. 

• Synergize in anti-viral response/IFN-p induction. 

• Neutrophil activation/ respiratory burst. 

• Endothelial cell activation 

• Toxicities noted when patients treated with TNF/IFN-y as anti-viral therapy 



WO 2004/003019 



PCT/GB2003/002804 



124 




• Fractalkine expression by human astrocytes. 




• Many papers on inflammatory responses - i.e. LPS, also macrophage 




activation. 




• Anti-TNF and anti-IFN-y synergize to protect mice from lethal 




endotoxemia. 



TGF-p/IL-1 


• Prostaglndin synthesis by osteoblasts 

• IL-6 production by intestinal epithelial cells (mflarnmation model) 

• Stimulates IL-1 1 and IL-6 in lung fibroblasts (inflammation model) 

• IL-6 and IL-8 production in the retina 


TGF-p/IL-6 


• Chondrocarcoma proliferation 


IL-l/IL-2 


• B-cell activation 

• LAK cell activation 

• T-cell activation 

• IL-1 synergy with IL-2 in the generation of lymphokine activated killer 
p^IIq i<2 mprliatpfl V>v TNF-alnha and beta ( 1 vrnnhotoxin^ . Cytokine. 1992 

Nov4(6V479-87 


TT 1 /TT 'X 




IL-l/IL-4 


• B-cell activation 

• IL-4 induces IL-1 expression in endothelial cell activation. 


TT 1 /TT C 

IL-l/IL-5 




IL-l/IL-6 


• B cell activation 

• T cell activation (can replace accessory cells) 

• IL-1 induces IL-o expression 

• C3 and serum amyloid expression (acute phase response) 

• HIV expression 

• Cartilage collagen breakdown. 


IL-l/IL-7 


• IL-7 is requisite for IL-1 -induced thymocyte proliferation. Involvement of 
IL-7 in the synergistic effects of granulocyte-macrophage colony- 
stimulating factor or tumor necrosis factor with IL-1. J Immunol. 1992 Jan 
1;148(1):99-105. 


IL-l/IL-8 




IL-l/IL-10 




IL-l/IL-11 


• Cytokines synergistically induce osteoclast differentiation: support by 
immortalized or normal calvarial cells. Am J Physiol Cell Physiol 2002 
Sep;283(3):C679-87. (Bone loss) 


IL-l/IL-16 


• Correlation of circulatmg mterleukm 16 with promflammatory cytokmes in 
patients with rheumatoid arthritis. Rheumatology (Oxford). 2001 
Apr;40(4):474~5. No abstract available. 


TT 1 /TT 1 7 


• TnTiih-itinn n-f inter1f»itlrin-1 7 nTPvent*? the development of arthritis in 

vaccinated mice challenged with Borrelia burgdorferi. Infect Immun. 2003 
Jun;71(6):3437-42. 

• Contribution of interleukin 17 to human cartilage degradation and synovial 
inflammation in osteoarthritis. Osteoarthritis Cartilage. 2002 
Oct;10(10):799-807. 

• Abstract Interleukin- 1, tumor necrosis factor alpha, and interleukin- 17 
synergistically up-regulate nitric oxide and prostaglandin E2 production in 
explants of human osteoarthritic knee menisci. Arthritis Rheum. 2001 
Sep;44(9):2078-83. 


IL-l/IL-18 


• Association of interleukin- 1 8 expression with enhanced levels of both 

mterleukin-lbeta and tumor necrosis factor alpha in knee synovial tissue of 
patients with rheumatoid arthritis. Arthritis Rheum. 2003 Feb;48(2):339-47. 


IL-l/IFN-g 




IL-2/IL-3 


• T-cell proliferation 

• B cell proliferation 



WO 2004/003019 



PCT/GB2003/002804 



125 



IL-2/IL-4 


• B-cell proliferation 

• T-cell proliferation 

• (selectively inducing activation of CD 8 and NK lymphocytes)IL-2R beta 
agonist Pl-30 acts in synergy with IL-2, IL-4, IL-9, and IL-15: biological 
and molecular effects. J Immunol 2000 Oct 15:165(8 1: 4312-8. 


IL-2/IL-5 


• B-cell proliferation/ Ig secretion 

• IL-5 induces IL-2 receptors on B-cells 


IL-2/IL-6 


• Development of cvtotoxic T- cells 


IL-2/IL-7 




IL-2/IL-9 


• See IL-2/IL-4 (NK-cells) 


IL-2/IL-10 


• B-cell activation 


IL-2/IL-12 


• x.l-iz synergizes witn xi^-z to rnauce lympnoKane-actrvatea cytotoxicity 
and perforin and granzyme gene expression in fresh human NK cells. Cell 
Immunol 1995 Oct l;165(l):33-43. (T-cell activation) 


IL-2/IL-15 


• See IL-2/IL-4 (NK cells) 

• (l cell activation and proliferation) IL-15 and IL-2: a matter of life and 
death for T cells in vivo. Nat Med. 2001 Jan;7(l): 114-8. 


IL-2/IL-16 


• Synergistic activation of CD4+ T cells by IL-16 and IL-2. J Immunol 1998 
JVlar I;loU(3):21 15-20. 


IL-2/IL-17 


• Evidence for the early involvement of interleukin 17 in human and 

experimental renal aiiogratt rejection. J JratnoL z002 Jul;197(3):322-32. 


IL-2/IL-18 


• interieuKin lo (ii^-io; in synergy with JLL~z induces lethal lung injury m 
mice: a potential role for cytokines, chemokines, and natural killer cells in 
the pathogenesis of interstitial pneumonia. Blood. 2002 Feb 15;99(4):1289- 
98. 


IL-2/TGF-R 

XJ— / X VJX kJ 


» v^uiiLiui ox eiiector rate, iransiorming growtn tactor beta 1 and 
nitt^i it/ ujviii z. ayiAcigiZiC lu picvciii dpopiosis and promote eiiector 
expansion JExvMed 1995 Ser> 1 -182nV69Q-7no 


IL-2/IFN-y 


• Ig secretion by B-cells 

• IL-2 induces IFN-y expression by T-cells 


TT -9/rFTVLrv/ft 


• rv one 


IL-3/IL-4 


• Synergize in mast cell growth 

• Synergistic effects of IL-4 and either GM-CSF or IL-3 on the induction of 
CD23 expression by human monocytes: regulatory effects of IFN-alpha and 
IFN-gamma. Cytokine. 1994 Jul;6(4):407-13. 


TT ^ /TT ^ 




IL-3/IL-6 




TT "2 /TT7VT *r 


• IL-4 and IFN-ganima synergistically increase total polymeric IgA receptor 
levels in human intestinal epithelial cells. Role of protein tyrosine kinases. 
J Immunol. 1996 Jun 15;156(12):4807-14. 


IL-3/GM-CSF 


• Differential regulation of human eosinophil IL-3, IL-5, and GM-CSF 
receptor alpna-cnain expression by cytokines: IL-3, IL-5, and GM-CSF 
ao wn-reguiate ijl-d receptor alpna expression witn loss oi IL-5 
responsiveness, out up-reguiate receptor alpna expression. J Immunol. 
2003 Jun l;170(ll):5359-66. (allergic inflammation) 


IL-4/IL-2 


• IL-4 svnereisticallv enhances both IL-2- and IL-12-indiiceH TFIM- i a p mm ^ \ 
expression in murine NK cells. Blood. 2003 Mar 13 TEpub ahead of print] 


IL-4/IL-5 


• Enhanced mast cell histamine etc. secretion in response to IgE 

• A Th2-like cytokine response is involved in bullous pemphigoid, the role of 
IL-4 and IL-5 in the pathogenesis of the disease. Int J Immunopathol 
Pharmacol 1999 May-Aug;12(2):55-61. 


IL-4/IL-6 




IL-4/IL-10 




IL-4/IL-11 


• Synergistic interactions between interleukin-1 1 and interlenkin-4 in support 
of proliferation of primitive hematopoietic progenitors of mice. Blood. 



WO 2004/003019 



PCT/GB2003/002804 



126 





1991 Sep 15;78(6):1448-51. 


IL-4/IL-12 

XI t VI i-J — ' X 


• S»vnprai«;tic effects of TL-4 and IL-18 on IL-12-deoendent IFN-eamma 
production by dendritic cells. J Immunol 2000 Jan 1;164(1):64-71. 
(increase Thl/Th2 differentiation) 

• TL-4 ^vnereisticallv enhances both IL-2- and IL-12-induced IFN-I gamma) 
expression in murine NK cells. Blood. 2003 Mar 13 fEpub ahead of print! 


IL-4/IL-13 


• Abstract Interleukin-4 and interleukin- 13 signaling connections maps. 
Sripnrr 200*3 Tun 6-300C5625V 1527-8 fallerev asthma") 

• Inhibition of the IL-4/IL-13 receptor system prevents allergic sensitization 
without affectincr established allerev in a mouse model for allergic asthma. 
J Allergy Clin Immunol. 2003 Jun;lll(6):1361-1369. 


IL-4/IL-16 


• (asthma) Interleukin (IL)-4/IL-9 and exogenous IL-16 induce IL-16 
production by BEAS-2B cells, a bronchial epithelial cell line. Cell 
Immunol. 2001 Feb l;207(2):75-80 


IL-4/IL-17 


• Interleukin (IL)-4 and IL-17 synergistically stimulate IL-6 secretion in 

liiiman nrtlrvnir* mvnfi Krn i%1 a <;tQ ftif 7 A/fnJ A/fpH 900'? Nnvl 0f5V6^ 1 -4 

UU.llJ.Cli.JL V^vJlvllllV/ 111 V Ulllsl vJ UlClO ID . XftL iv JVJ.L/1 JriCU. Z-W l'lUVjXVlJ y.Ww' 1 t . 

(Gut inflairimation) 


TT A/TT 94 


a TT OA ic a-vTAVf^c c/=>r1 V»A/ r rat unr? fnimnn mo r»TT\T^Vl n Cf^c TtYHYilltl (thi rhCr\) 0000 
• J.J_/ — Z-T" xo CA.piCbbCLl Uy LcLV cilXKX HU.llla.ll lllav/1 wjJlldgt'S. xfilffllAIl\JlJl>Vl\jx< i y. £*\J\J 

Jul;205(3):321-34. 


TT 4/TT -0*i 


« AKc+rcn-t TsTrtx/' TT _17 fnmilv mpmliprQ nrnmntp TVi! nt Th'? TPWrni^p*? in the 

• ADa IXdv/L 1NCW 1J—/ 1 / ldlllliy lliciliut/i o ljx VJ ixivJ LV/ jlj.ii \jx nix ivvojjvJxxot/O xia nx^ 

lung: in vivo function of the novel cytokine IL-25. J Immunol. 2002 Jul 
l;169(l):443-53. (allergic inflammation) 

• Abstract Mast cells produce interleukin-25 upon Fcepsilon Rl-mediated 
activation. Blood. 2003 May l;101(9):3594-6. Epub 2003 Jan 02. (allergic 

1 Tl fl £» TY1TY1 54 "H C\Yl 1 

IJLlllallllllaLlAJll y 


TT /I /TT7XT 

IL-4/lFN-y 


• ADSiract inTeneuKin *» niQuces micricuKin o protiuuLiun oy ciiuuuiciiai uciio. 
synergy with interferon-gamma. Eur J Immunol. 1 99 1 Jan;2 1 ( 1 ) : 97- 101. 


IL-4/SCF 


• Regulation of human intestinal mast cells by stem cell factor and IL-4. 
Immunol jctev. zuui rcu,i / y.D i -ou. xveview. 


IL-5/IL-3 


• Differential regulation of human eosinophil IL-3 5 IL-5, and GM-CSF 
receptor alpha-chain expression by cytokines: IL-3, IL-5, and GM-CSF 

A r\-»im rprm1(j+p TT ^ T"<=»r**=»-r\fr»T cilnrm pynrpccinti ■\x/it"Ti IncG r\~f TT 
CIO WIl-lCgUlclLC 11j J ICv/CpiUl dlJJlla CA.piCoolvJll W1LUL IvJoo KJX Xl—i~*J 

responsiveness, but up-regulate IL-3 receptor alpha expression. J Immunol. 
2003 Jun l;170(ll):5359-66. (Allergic inflammation see abstract) 


IL-5/IL-6 




IL-5/IL-13 


• Inhibition of allergic airways inflammation and airway 

hyperresponsiveness in mice by dexamethasone: role of eosinophils, IL-5, 
entaxin andTL-13 J Allergy Clin Immunol 2003 Mav*lll(5V 1049-61. 


TT -S/TT -1 7 

XX-i~^J! XX-J X 1 


• TntprlenVin-1 7 nrehe^trates the eranulocvte influx into airwavs after 

allergen inhalation in a mouse model of allergic asthma. Am J Respir Cell 
MolBiol. 2003 Jan;28(l):42-50. 


IL-5/IL-25 


• Abstract New IL-17 family members promote Thl or Th2 responses in the 
lung: in vivo function of the novel cytokine IL-25. J Immunol. 2002 Jul 

1 *1 M(\ \ -44*3 -5 3 (alleroic inflarnmation^ 
m ATncfrjmt A/fnct pplk •nrndnre inter1enVin-'75 linmi Fcen<iilnn RT-mediated 

• ^j.L?OLld^/L lVlCXO L V^-Cllo L/l vJLXLX^/t' JJLXLWJ.XwLlXVJ.XX a<J U.JL/UXX X vwUOXlUll XVI. lUvUlUlUU 

activation Blood 2003 Mav 1-101C9V3594-6 Eoub 2003 Jan 02. (allergic 
inflammation) 


IL-5/IFN-y 




IL-5/GM-CSF 


• Differential regulation of human eosinophil IL-3, IL-5, and GM-CSF 
receptor alpha-chain expression by cytokines: IL-3, IL-5, and GM-CSF 
down-regulate IL-5 receptor alpha expression with loss of IL-5 
responsiveness, but up-regulate IL-3 receptor alpha expression. J Immunol. 
2003 Jun l;170(ll):5359-66. (Allergic inflammation) 


IL-6/IL-10 




IL-6/IL-11 




IL-6/IL-16 


• Interleukin-16 stimulates the expression and production of pro- 



WO 2004/003019 



PCT/GB2003/002804 



127 





inflammatory cytokines by human monocytes. Immunology. 2000 


IL-6/IL-17 


• Stimulation of airway mucin gene expression by interleukin (IL)-17 

fhrmio-Ti TT f\ ■nararrmp/aiitocrine loon J Hinl CIipwi 2003 lVfav 

9;278(19): 17036-43. Epub 2003 Mar 06. (airway inflammation, asthma) 


IL-6/IL-19 


• Abstract IL-19 induces production of IL-6 and TNF-alpha and results in 

~p.ii nr*n-ntrkcic fhrrmoh TNTF-alrVha / Tmmunnl 900? Oct 1 5-1 69fRV42R8- 

97. 


TT £/TTh"NJ er 




IL-7/IL-2 


• Interleukin 7 worsens graft-versus-host disease. Blood. 2002 Oct 
l;100(7):2642-9. 


EL-7/IL-12 


• Synergistic effects of IL-7 and IL-12 on human T cell activation. J 
Immunol 1995 May 15;154(10):5093-102. 


IL-7/IL-15 


• Interleukin-7 and interleukin- 15 regulate the expression of the bcl-2 and c- 
myb genes m cutaneous 1-celi lymphoma cells, rSlooa. zuui jnov 
l;98(9):2778-83. (growth factor) 


IL-8/IL-11 


• Abnormal production of interleukin (IL)-1 1 and IL-8 in polycythaemia 
vera. Cytokine. 2002 Nov 21 ;20(4): 178-83. 


IL-8/IL-17 


• The Role of IL-17 in Joint Destruction. Drug News Per sped. 2002 
Jan;15(l): 17-23. (arthritis) 

• Abstract Interleukin- 17 stimulates the expression of interleukin- 8, growth- 
related oncogene-alpha, and granulocyte-colony-stimulating factor by 
human airway epithelial cells. Am J Respir Cell Mo I Biol. 2002 
Jun;Zo(o): /4o-jo. (airway inriammationj 


IL-8/GSF 


• Interleukin- 8: an autocrine/paracrine growth factor for human 

hematopoietic progenitors acting in synergy with colony stimulating factor- 
1 to promote monocyte-macrophage growth and differentiation. Exp 
Hematol 1999 Jan;27(l):28-36. 


IL-8/VGEF 


• Intracavitary VEGF. bFGF, IL-8, IL-12 levels in primary and recurrent 
malignant glioma. JNeurooncol. 2003 May;62 (3) .297-303. 


IL-9/IL-4 


• Anti-mterleukm-9 antibody treatment inhibits airway lntlammation and 
hyperreactivity in mouse asthma model. Am J Respir Crit Care Med. 2002 
Aug 1;166(3):409-16. 


IL-9/IL-5 


• Pulmonary overexpression of IL-9 induces Th2 cytokine expression, 
leading to immune pathology. J Clin Invest. 2002 Jan;109(l):29-39. 

• Th2 cytokines and asthma. Interleukin-9 as a therapeutic target for asthma. 
Respir Res. 2001;2(2):o0-4. iipubzUUl reb Id. Keview. 

• Abstract mterieuKin-y ennances lnieneuKm- d receptor exprebbion, 
differentiation, and survival of human eosinophils. Blood. 2000 Sep 
15;96(6):2 163-71 (asthma) 


IL-9/IL-13 


• Anti-interleukin-9 antibody treatment inhibits airway inflammation and 
nyperreacnvity m mouse astnma moaei. yy.7?z »/ itespu i^rii K^are lvieu, 
Aug i,ioo(3j.4uy-io. 

• Direct effects of interleukin- 13 on epithelial cells cause airway 
hyperreactivity and mucus overproduction in asthma. Nat Med. 2002 

A.Ug,o^o j.OOJ-7. 


IL-9/IL-16 


• See IL-4/IL-16 


IL-10/IL-2 


• The interplay of interleukin- 10 (IL-10) and interleukin-2 (IL-2) in humoral 
immune responses: IL-10 synergizes with IL-2 to enhance responses of 
human B lymphocytes in a mechanism which is different from upregulation 
of CD25 expression. Cell Immunol 1994 Sep;157(2):478-88. 


IL-10/IL-12 




IL-10/TGF-P 


• IL-10 and TGF-beta cooperate in the regulatory T cell response to mucosal 
allergens in normal immunity and specific immunotherapy. Eur J 
Immunol 2003 May;33(5): 1205-14. 


IL-10/IFN-y 





WO 2004/003019 



PCT/GB2003/002804 



128 



IL-ll/IL-6 


• Interleukin-6 and interleukin-1 1 support human osteoclast formation by a 
RANKL-mdependent mechanism. Bone, 2003 Jan;32(l):l-7. (bone 
resorption in inflammation) 


IL-ll/IL-17 


• Polarized in vivo expression of IL-1 1 and IL-17 between acute and chronic 
skin lesions. J Allergy Clin Immunol 2003 Apr;lll(4):875-81. (allergic 
dermatitis i 

ytvx Axm nuu r 

• IL-1 7 promotes bone erosion in murine collagen-induced arthritis through 
loss of the receptor activator of NF-kappa B ligand/osteoprotegerin 
balance. J Immunol. 2003 Mar l;170(5):2655-62. 


IL-11/TGF-P 


• Polarized in vivo expression of IL-1 1 and IL-1 7 between acute and chronic 
skin lesions. J Allergy Clin Immunol 2003 Apr;lll(4):875-81. (allergic 
dermatitis) 


IL-12/IL-13 


• Relationship of Interleukin-1 2 and Interleukin-1 3 imbalance with class- 
specific rheumatoid factors and anticardiolipin antibodies in systemic lupus 
erythematosus. Clin Rheumatol 2003 May;22(2): 107-11. 


IL-12/IL-17 


• Upregulation of interleukin- 1 2 and - 1 7 in active inflammatory bowel 
disease Scand J Gastroenterol 2003 Feb ;38(2): 180-5. 


IL-12/IL-18 


• Synergistic proliferation and activation of natural killer cells by interleukin 
12 and interleukin 18. Cytokine. 1999 Nov;ll(l l):822-30. 

• Inflammatory Liver Steatosis Caused bv IL-1 2 and IL-1 8. J Interferon 
Cytokine Res. 2003 Mar;23(3): 155-62. 


TT -1 ?/TT 


• nter1eiikm»23 rather than interleukin- 12 is the critical cvtokine for 
autoimmune mflammation of the brain. Nature. 2003 Feb 
13;421(6924):744-8. 

• Abstract A unique role for IL-23 in promoting cellular immunity. J Leukoc 
Biol 2003 Jan;73(l):49-56. Review. 


IL-12/IL-27 


• Abstract IL-27, a heterodimeric cytokine composed of EBB and p28 
protein, induces proliferation of naive CD4(+) T cells. Immunity. 2002 
Jun:16f6):779-90. 


IL-12/IFN-y 


• IL-12 induces IFN-y expression by B and T-cells as part of immune 
stimulation. 


TT 1 ^/TT 


• See TT -5/IL-13 


IL-13/IL-25 


• Abstract New IL-1 7 family members promote Thl or Th2 responses in the 
lung: in vivo function of the novel cytokine IL-25. J Immunol. 2002 Jul 

1 •! 69(1 V443-53. (allereic inflammation) 

• Abstract Mast cells produce interleukin-25 upon Fcepsilon Rl-mediated 
activation. Blood. 2003 May l;101(9):3594-6. Epub 2003 Jan 02. (allergic 
inflammation) 


IL-15/IL-13 


• Differential expression of interleukins (IL)-13 and IL-1 5 in ectopic and 
eutopic endometrium of women with endometriosis and normal fertile 
women. Am JReprod Immunol. 2003 Feb;49(2):75-83. 


IL-15/IL-16 


• IL-1 5 and IL-1 6 overexpression in cutaneous T-cell lymphomas: stage- 
dependent increase in mycosis fungoides progression. Exp Dermatol. 2000 
Aug;9(4):248-51. 


IL-15/IL-17 


• Abstract IL-1 7, produced by lymphocytes and neutrophils, is necessary for 
litDopolvsaccharide-induced airway neutrophilia: IL-1 5 as a possible trigger. 
J Immunol 2003 Feb 15;170(4):2106-12. (airway inflammation) 


IL-15/IL-21 


• IL-2 1 in Synergy with IL-1 5 or IL-1 8 Enhances IFN-gamma Production in 
Human NK and T Cells. J Immunol 2003 Jun l;170(ll):5464-9. 


IL-17/IL-23 


• Interleukin-23 promotes a distinct CD4 T cell activation state characterized 
by the production of interleukin- 17. JBiol Chem. 2003 Jan 
17;278(3):1910-4. Epub 2002 Nov 03 


IL-17/TGF-p 


• Polarized in vivo expression of IL-1 1 and IL-1 7 between acute and chronic 
skin lesions. J Allergy Clin Immunol 2003 Apr;ll 1(4): 875-81. (allergic 
dermatitis) 


IL-18/IL-12 


• Synergistic proliferation and activation of natural killer cells by interleukin 



WO 2004/003019 



PCT/GB2003/002804 



129 





12 and interleukin 18. Cytokine. 1999 Nov;ll(l l):822-30. 
• Abstract Inhibition of in vitro immunoglobulin production by IL- 1 2 in 
murine chronic graft-vs.-host disease: synergism with IL-18. Eur J 
Immunol 1998 Jun;28(6):20 17-24. 


IL-18/IL-21 


• IL-21 in Synergy with IL-15 or IL-18 Enhances IFN-gamma Production in 
Human NK and T Cells. J Immunol 2003 Jun 1;170(1 1):5464-9. 


IL-18/TGF-p 


• Interleukin 1 8 and transforming growth factor betal in the serum of 
patients with Graves' ophthalmopathy treated with corticosteroids. Int 
Immunopharmacol 2003 Apr;3(4): 549-52. 


IL-18/IFN-Y 




Anti-TNF 
ALPHA/anti-CD4 


• Synergistic therapeutic effect in DBA/1 arthritic mice. 



WO 2004/003019 PCT/GB2003/002804 

130 

Annex 3: Oncology combinations 



Target 


Disease 


Pair with 


CD89* 


Use as cytotoxic cell recruiter 


all 








CD19 


B cell lvnTDhotnas 

J-* V/VJi AY A.AJ.Jw'AAVo'AAAtliD 










HLA-DR 


B cell Ivninhcvnia^ 

J— ' vUJll A V AllLJAAvrAlAClO 














v^xy„? 


CD38 


A/Til 1 tl^l f» YYY\Te*1i~\YV1Cl 

ivmiLipic my civ ilia 














XXJ_(/\-X-/XV 


CD138 


lVTultinle mvplnma 

At AtAALAJL/AVs AAA Y \s -LVJ LXiCL 


CTnR 

V^X-/ J) o 












XXX_/xjl. XvXV 


CD138 


T IITIO" PflTlPPr 

X_/UXXg l/dXXOC/X 








CEA 


CD33 


A pnfp Tmvplnrl 1 ^rmr^Vi nm^ 
*VW UXG lllyCHJVl IJ'IIaJPIaIJIIacI 


V^X^O^T 






XX JLj/\~ J_J X\- 


CD56 


Lixnf* cancer 

A—/ UAlfi vsUAAV/wA 


V^X^ 1 JO 






CFA 


CEA 


Pan carrtnoma 

A ClAA v/iiA v^AAAUAAAu- 


ivxj-i x icocptoi 


VEGF 

V J / V — 1 A. 


P qt) rairin/ima 

X dXX OCXXOiAiUlAACt 


ivldx receptor 


VFGF 


TX rj /"» Q •*•/"• 1 "M <""vt"V1 id 

x dii udrcinoijia 


iVLbl receptor 


A V/ O V^IJ I.AJ 1 






IL-13 


A <5rliTVi5?/n'i'ilTnfYnflT"i7 
■Ti-ij ixiixxci/ jj uxxxiuilaX y 






inflamma iinn 

AAAAAUAA AAAXCA llvll 


TT _S 






X_i vJ Id A.JXXI O 1 






MDC 






TARC 












IL-9 

lAv 7 






EGFR 

A_/\ IX AX. 






CD40L 






IL-25 






MCP-l 






TGFR 

X VJX IJ 


IL-4 


Asthina 


IX/ A «J> 






IL-5 

A J — l w/ 






Eo taxing s^ 






MDC 






TARC 






TNFc£ 






TT -Q 






EGFR 






CD40L 






IL-25 






MCP-l 






TGFP 


Eotaxin 


Asthma 


IL-5 






Eotaxin-2 






Eotaxin-3 


EGFR 


cancer 1 


HER2/neu 






HER3 






HER4 


HER2 


cancer 


HER3 



WO 2004/003019 



PCT/GB2003/002804 



131 



TNFR1 



RA/Crohn's disease 



HER4 
IL-1R 
IL-6R 



TNFa 


RA/Crohn's disease 


IL-la/0 






IL-6 






IL-18 






ICAM-1 






IL-15 






IL-17 


IL-1R 


RA/Crohn's disease 


IL-6R 






IL-18R 


IL-18R 


RA/Crohn's disease 


IL-6R 



WO 2004/003019 



132 



PCT/GB2003/002804 



S-i 

a 

GO 



"3 

S3 

on 
OS 



CD 

a 



a 
op 

o 



io 

p 



o 
o 
to 



a, 
o 
o 



o 
o 
*o 



s 

o 



C/3 



s 

Q 



a 

o 



on 
< 



a 
§ 



H 



o 



io 

IT 




CN O 

T— I 1— 

II II 




Oto «o io 
.III 

— T-H t— I 1— I 



oo 

II 

to 

*o 

Os 
1 — I 
I 

*o 

I 



li II 

^ w 

□ o 

ON ON 



OO CO 

IE II 

CJ U 

rS co 

o o 

0\ ON 



to to »o to 



H H H H 



X 
i — i 

o 



to 



§ 

1 

1 — I 

< 



J— < 

§ 



H 

1/3 



o 



O K> 

'■5 s 



o 
o 



.11 

5S CI 



to *7 
o O 



E 

Oh 



O 



S3 

cr 



1=3 
© 



O 

o 

CO 



X 

CO 

^io 



o 
io 



o 

g 

CO 



s 
§ 



3 



a 
§ 

1 



o 

co 



on 



to 



i 

to 

I 



i 

o 





to 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



133 



PCT/GB2003/002804 



o 



CD 



o 
o 



c O 

CD 

CO 

^> ^3 Tj 
Q io *o *o *o 



ro >o m m 
II II II II 



H H H H 



o o 

o o 

II II 

to *n 

i i 



o 
m 

II 

1 w 

5 P 

-8 : 



o 
o 



II 

8 

CO 

O 
i 



o 



Q 



O 
O 



o 
o 



o 
o 



=3. 

o 



s § 

o , " H 

o ^ 

o c5 

> <+-c 




§ 






i 

I* 





1 

o 
o 



O 



< 

GO 
i 



on 

1 



3 1 



CO 



CN 
GO 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



134 



PCT/GB2003/002804 



Claims 

1 . A dual-specific ligand comprising a first immunoglobulin single variable 
domain having a binding specificity to a first epitope or antigen and a second 
complementary immunoglobulin single variable domain having a binding activity 
to a second epitope or antigen, wherein one or both of said antigens or epitopes 
acts to increase the half-life of the ligand in vivo and wherein said first and second 
domains lack mutually complementary domains which share the same specificity, 
provided that said dual specific ligand does not consist of an anti-HS A Vh domain 
and an anti-p galactosidase V K domain. 

2. A dual-specific ligand according to claim 1, comprising at least one single 
heavy chain variable domain of an antibody and one complementary single light 
chain variable domain of an antibody such that the two regions are capable of 
associating to form a complementary VH/VL pair. 

3 . A dual specific ligand according to claim 2 wherein the Vh and Vl are 
provided by an antibody scFv fragment. 

4. A dual-specific ligand according to claim 2 wherein the Vh and Vl are 
provided by an antibody Fab region. 

5. A four chain IgG immunoglobulin ligand comprising a dual specific 
ligand of claim 2. 

6. A four chain IgG immunoglobulin ligand according to claim 5, wherein 
said IgG comprises two dual specific ligands, said dual specific ligands being 
identical in their variable domains. 

7. A four chain IgG immunoglobulin ligand according to claim 5, wherein 
said IgG comprises two dual specific ligands, said dual specific ligands being 
different in their variable domains. 

8. A ligand comprising a first immunoglobulin variable domain having a first 
antigen or epitope binding specificity and a second immunoglobulin variable 
domain having a second antigen or epitope binding specificity wherein one or 
both of said first and second variable domains bind to an antigen which increases 
the half-life of the ligand in vivo, and the variable domains are not complementary 
to one another. 

9. A ligand according to claim 8 wherein the first and the second 
immunoglobulin variable domains are heavy chain variable domains (Vh). 

10. A ligand according to claim 8 wherein the first and the second 
immunoglobulin variable domains are light chain variable domains (Vl). 

11. A ligand according to claim any preceding claim, wherein the first and 
second epitopes bind independently, such that the dual specific ligand may 
simultaneously bind both the first and second epitopes or antigens. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



135 



PCT/GB2003/002804 



12. A ligand according to claim 1 1 ? wherein the dual specific ligand comprises 
a first form and a second form in equilibrium in solution, wherein both epitopes or 
antigens bind to the first form independently but compete for binding to the 
second form. 

13. A ligand according to any preceding claim wherein the variable regions 
are derived from immunoglobulins directed against said epitopes or antigens. 

14. A ligand according to any preceding claim, wherein said first and second 
epitopes are present on separate antigens. 

15. A ligand according to any one of claims 1 to 1 1 5 wherein said first and 
second epitopes are present on the same antigen. 

16. A ligand according to any preceding claim comprising a variable domain 
that is derived from a repertoire of single antibody domains. 

17. A ligand of claim 16 wherein said repertoire is displayed on the surface of 
filamentous bacteriophage and wherein the single antibody domains are selected 
by binding of the bacteriophage repertoire to antigen. 

18. A ligand according to any preceding claim wherein the sequence of at 
least one variable domain is modified by mutation or DNA shuffling. 

19. A dual-specific ligand according to any preceding claim wherein the 
variable regions are non-covalently associated. 

20. A dual-specific ligand according to any one of claims 1 to 18 wherein the 
variable regions are covalently associated. 

21. A dual-specific ligand according to claim 20 wherein the covalent 
association is mediated by disulphide bonds. 

22. A dAb monomer ligand specific for TNFa, which dissociates from human 
TNFa with a dissociation constant (Kd) of 50nM to 20pM, and a K 0 ff rate constant 
of SxlO" 1 to lxlO" 7 s** 1 , as determined by surface plasmon resonance. 

23. A dAb monomer ligand specific for TNFa according to claim 22, wherein 
the dAb is a Vk. 

24. A dAb monomer ligand specific for TNF receptor 1 (p55), which 
dissociates from human TNF receptor 1 with a dissociation constant (Kd) of 50nM 
to 20pM, and a K 0 ff rate constant of 5x1 0" 1 to lxl 0" 7 s" 1 , as determined by surface 
plasmon resonance. 

25. A dAb monomer ligand according to claim 22 to claim 24, wherein the 
monomer neutralises human TNFa or TNF receptor 1 in a standard cell assay 
with an ND50 of 500nM to 50pM. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



136 



PCT/GB2003/002804 



26. A dAb monomer ligand specific for TNF receptor 1 (p55), wherein the 
dAb antagonises the activity of the TNF receptor 1 in a standard cell assay with 
an ND 50 of <100nM, and at a concentration of <10pM the dAb agonises the 
activity of the TNF receptor 1 by <5% in the assay. 

27. A dAb monomer ligand specific for serum albumin (SA) which dissociates 
from SA with a dissociation constant (K d ) of InM to 500jjM ? as determined by 
surface plasmon resonance. 

28. A dAb monomer ligand according to claim 27, wherein the monomer 
binds SA in a standard ligand binding assay with an IC50 of InM to SOOjjM. 

29. A dAb monomer ligand specific for TNFa, wherein the dAb comprises 
the amino acid sequence of TAR1-5-19 or a sequence that is at least 80% 
homologous thereto. 

30. A dAb monomer ligand specific for TNFa, wherein the dAb comprises 
the amino acid sequence of TAR1-5 or a sequence that is at least 80% 
homologous thereto. 

31. A dAb monomer ligand specific for TNFa, wherein the dAb comprises 
the amino acid sequence of TAR1-27 or a sequence that is at least 80% 
homologous thereto. 

32. A dAb monomer ligand specific for TNF receptor 1 ? wherein the dAb 
comprises the amino acid sequence of TAR2-10 or a sequence that is at least 80% 
homologous thereto. 

33. A dAb monomer ligand specific for TNF receptor 1, wherein the dAb 
comprises the amino acid sequence of TAR2-10 or a sequence that is at least 90% 
homologous thereto. 

34. A dAb monomer ligand specific for TNF receptor 1 , wherein the dAb 
comprises the amino acid sequence of TAR2-5 or a sequence that is at least 80% 
homologous thereto. 

35. A dAb monomer ligand specific for TNF receptor 1 , wherein the dAb 
comprises the amino acid sequence of TAR2-5 or a sequence that is at least 90% 
homologous thereto. 

36. A dAb monomer ligand specific for SA, wherein the dAb comprises the 
amino acid sequence of MSA-16 or a sequence that is at least 80% homologous 
thereto. 

37. A dAb monomer ligand specific for SA, wherein the dAb comprises the 
amino acid sequence of MSA-26 or a sequence that is at least 80% homologous 
thereto. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



137 

38. A dAb monomer according to any one of claims 29 to 37, wherein the 
TNFoc, TNF receptor 1 or SA is in human form. 

39. A dAb monomer further comprising a terminal Cys residue. 

40. A dAb monomer according to any one of claims 29 to 38, further 
comprising a terminal Cys residue. 

41. A dual specific ligand comprising at least one dAb monomer according to 
any one of claims 22 to 40. 

42. A dual specific ligand according to claim 41 , which is a dimer. 

43. A dual specific ligand according to claim 42, wherein the dimer comprises 
anti-human TNF alpha dAb according to any one of claims 22, 23 and 28-30, and 
an anti-SA dAb according to any one of claims 26, 27 and 34-36. 

44. A dual specific ligand according to claim 42, wherein the dimer is a 
homo- or hetero-dimer comprising first and second anti-human TNF alpha dAbs, 
each dAb being according to any one of claims 22, 23 and 28-30. 

45. A dual specific ligand according to claim 41, which is a trimer. 

46. A dual specific ligand according to claim 45, which is a homotrimer 
comprising three copies of an anti-human TNF alpha dAb according to any one of 
claims 22, 23 and 29-3 1 . 

47. A ligand according to any preceding claim, which comprises a universal 
framework. 

48. A ligand according to claim 47, wherein the universal framework 
comprises a Vh framework selected from the group consisting of DP47, DP45 and 
DP38; and/or the V L framework is DPK9. 

49. A ligand according to any preceding claim which comprises a binding site 
for a generic ligand. 

50. The ligand of claim 49, wherein the generic ligand binding site is selected 
from the group consisting of protein A, protein L and protein G. 

51. A ligand according to any preceding claim, wherein the ligand comprises a 
variable domain having one or more framework regions comprising an amino acid 
sequence that is the same as the amino acid sequence of a corresponding 
framework region encoded by a human germline antibody gene segment, or the 
amino acid sequences of one or more of said framework regions collectively 
comprises up to 5 amino acid differences relative to the amino acid sequence of 
said corresponding framework region encoded by a human germline antibody 
gene segment. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



138 



PCT/GB2003/002804 



52. A ligand according to any one of claims 1 to 51, wherein the ligand 
comprises a variable domain, wherein the amino acid sequences of FW1, FW2, 
FW3 and FW4 are the same as the amino acid sequences of corresponding 
framework regions encoded by a human germline antibody gene segment, or the 
amino acid sequences of FW1, FW2 ? FW3 and FW4 collectively contain up to 10 
amino acid differences relative to the amino acid sequences of corresponding 
framework regions encoded by said human germline antibody gene segment. 

53. The ligand according to claim 51 or claim 52, which comprises an 
antibody variable domain comprising FW1, FW2 and FW3 regions, and the 
amino acid sequence of said FW1, FW2 and FW3 are the same as the amino acid 
sequences of corresponding framework regions encoded by human germline 
antibody gene segments. 

54. The ligand according to any one of claims 51 to 53, wherein said human 
germline antibody gene segment is selected from the group consisting of DP47, 
DP45,DP48and DPK9. 

55. A ligand according to any preceding claim, comprising a Vh domain that 
is not a Camelid immunoglobulin variable domain. 

56. The ligand of Claim 55, comprising a Vh domain that does not contain one 
or more amino acids that are specific to Camelid immunoglobulin variable 
domains as compared to human Vh domains. 

57. A method for producing a ligand comprising a first immunoglobulin single 
variable domain having a first binding specificity and a second single 
immunoglobulin single variable domain having a second binding specificity, one 
or both of the binding specificities being specific for a protein which increases the 

half-life of the ligand in v/vo, the method comprising the steps of: 

(a) selecting a first variable domain by its ability to bind to a first epitope, 

(b) selecting a second variable region by its ability to bind to a second 
epitope, 

(c) combining the variable regions; and 

(d) selecting the ligand by its ability to bind to said first and second epitopes; 
wherein, when said variable domains are complementary, neither of said domains 
is a Vh domain specific for HSA. 

58. A method according to claim 57 wherein said first variable domain is 
selected for binding to said first epitope in absence of a complementary variable 
domain. 

59. A method according to claim 57 wherein said first variable domain is 
selected for binding to said first epitope in the presence of a third complementary 
variable domain in which said third variable domain is different from said second 
variable domain. 

60. Nucleic acid encoding a dual-specific ligand according to any one of 
claims 1 to 56. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

139 

61 . A nucleic acid according to claim 60 which is specific for TNFa, 
comprising the nucleic acid sequence of TAR1-5-19 or a sequence that is at least 
70% homologous thereto. 

62. A nucleic acid according to claim 60 which is specific for TNFa, 
comprising the nucleic acid sequence of TAR1-5 or a sequence that is at least 
70% homologous thereto. 

63. A nucleic acid according to claim 60 which is specific for TNFa, 
comprising the nucleic acid sequence of TAR1-27 or a sequence that is at least 
70% homologous thereto. 

64. A nucleic acid according to claim 60 which is specific for TNF receptor 1 , 
comprising the nucleic acid sequence of TAR2-10 or a sequence that is at least 
70% homologous thereto. 

65. A nucleic acid according to claim 60 which is specific for TNF receptor 1, 
comprising the nucleic acid sequence of TAR2-10 or a sequence that is at least 
80% homologous thereto. 

66. A nucleic acid according to claim 60 which is specific for TNF receptor 1 5 
comprising the nucleic acid sequence of TAR2h-5 or a sequence that is at least 
70% homologous thereto. 

67. A nucleic acid according to claim 60 which is specific for TNF receptor 1, 
comprising the nucleic acid sequence of TAR2h-5 or a sequence that is at least 
80% homologous thereto. 

68. A nucleic acid according to claim 60 which is specific for SA, comprising 
the nucleic acid sequence of MSA- 16 or a sequence that is at least 70% 
homologous thereto. 

69. A nucleic acid according to claim 60 which is specific for SA, comprising 
the nucleic acid sequence of MSA-26 or a sequence that is at least 70% 
homologous thereto. 

70. A vector comprising nucleic acid according to any one of claims 60 to 69. 

71 . A vector according to claim 70, further comprising components necessary 
for the expression of a dual-specific ligand. 

72. A host cell transfected with a vector according to claim 71 . 

73 . A method for producing a closed conformation multi-specific ligand 
comprising a first single epitope binding domain having a first epitope binding 
specificity and a non-complementary second epitope binding domain having a 
second epitope binding specificity, wherein the first and second binding 
specificities are capable of competing for epitope binding such that the closed 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



140 

conformation multi-specific ligand may not bind both epitopes simultaneously, 
said method comprising the steps of: 

a) selecting a first epitope binding domain by its ability to bind to a first 
epitope, 

b) selecting a second epitope binding domain by its ability to bind to a 
second epitope, 

c) combining the epitope binding domains such that the domains are in a 
closed conformation; and 

d) selecting the closed conformation multispecific ligand by its ability to bind 
to said first second epitope and said second epitope, but not to both said first and 
second epitopes simultaneously. 

74. A method according to claim 73 wherein the first and the second epitope 
binding domains are immunoglobulin variable heavy chain domains (v H )- 

75. A method according to claim 73 wherein the first and the second 
immunoglobulin variable domains are immunoglobulin variable light chain 
domains (Vl)- 

76. A method according to any one of claims 73 to 75 wherein the 
immunoglobulin domains are derived from immunoglobulins directed against said 
epitopes. 

77. A method according to any one of claims 73 to 76, wherein said first and 
second epitopes are present on separate antigens. 

78. A method according to any one of claims 73 to 76, wherein said first and 
second epitopes are present on the same antigen. 

79. A method according to any one of claims 73 to 78 wherein the variable 
domain is derived from a repertoire of single antibody domains. 

80. A method of claim 79 wherein said repertoire is displayed on the surface 
of filamentous bacteriophage and wherein the single antibody domains are 
selected by binding of the bacteriophage repertoire to antigen. 

81. A method of any one of claims 73 to 80 wherein the sequence of at least 
one immunoglobulin variable domain is modified by mutation or DNA shuffling. 

82. A closed conformation multispecific ligand comprising a first epitope 
binding domain having a first epitope binding specificity and a non- 
complementary second epitope binding domain having a second epitope binding 
specificity wherein the first and second binding specificities are capable of 
competing for epitope binding such that the closed conformation multi-specific 
ligand cannot bind both epitopes simultaneously. 

83. A closed conformation multispecific ligand according to claim 82, 
obtainable by a method according to any one of claims 73 to 80. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



141 



PCT/GB2003/002804 



84. A closed conformation multispecific ligand according to claim 82 or claim 
83, comprising more than one single heavy chain variable domain of an antibody 
or more than one light chain variable domain of an antibody. 

85. A closed conformation multi-specific ligand according to claim 84 
wherein the Vh y L are linked by a peptide linker. 

86. A closed conformation multi-specific ligand according to claim 84 
wherein the Vh or Vl are provided by an antibody Fab-like region. 

87. A closed conformation multi-specific ligand according to any one of 
claims 82 to 84 wherein the variable regions are non-covalently associated. 

88. A closed conformation multi-specific ligand according to any one of 
claims 82 to 84 wherein the variable regions are covalently associated. 

89. A closed conformation multi-specific ligand according to claim 87 
wherein the covalent association is mediated by disulphide bonds. 

90. A closed conformation multi-specific ligand according to any of claims 82 
to 89 which comprises a universal framework. 

91 . A closed conformation multi-specific ligand according to any of claims 82 
to 90 which comprises a binding site for a generic ligand. 

92. The closed conformation multi-specific ligand of claim 9 1 , wherein the 
generic ligand binding site is selected from the group consisting of protein A, 
protein L and protein G. 

93. A closed conformation ligand according to any of claims 82 to 92, 
wherein the ligand comprises a variable domain having one or more framework 
regions comprising an amino acid sequence that is the same as the amino acid 
sequence of a corresponding framework region encoded by a human germline 
antibody gene segment, or the amino acid sequences of one or more of said 
framework regions collectively comprises up to 5 amino acid differences relative 
to the amino acid sequence of said corresponding framework region encoded by a 
human germline antibody gene segment. 

94. The closed conformation ligand according to claim 93, wherein the ligand 
comprises a variable domain wherein the amino acid sequences of FW1, FW2, 
FW3 and FW4 are the same as the amino acid sequences of corresponding 
framework regions encoded by a human germline antibody gene segment, or the 
amino acid sequences of FW1, FW2, FW3 and FW4 collectively contain up to 10 
amino acid differences relative to the amino acid sequences of corresponding 
framework regions encoded by said human germline antibody gene segment. 

95. The closed conformation ligand according to claim 93 or claim 94, which 
comprises an antibody variable domain comprising FW1, FW2 and FW3 regions, 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



PCT/GB2003/002804 



142 

and the amino acid sequence of said FW1, FW2 and FW3 are the same as the 
amino acid sequences of corresponding framework regions encoded by human 
germline antibody gene segments. 

96. The closed conformation ligand according to any one of claims 92 to 95, 
wherein said human germline antibody gene segment is selected from the group 
consisting of DP47, DP45, DP48 and DPK9. 

97. A closed conformation ligand according to any one of claims 92 to 96, 
comprising a V H domain that is not a Camelid immunoglobulin variable domain. 

98. The closed conformation ligand of Claim 97, wherein the V H domain does 
not contain one or more amino acids that are specific to Camelid immunoglobulin 
variable domains as compared to human Vh domains. 

99. A closed conformation multi-specific ligand according to any one of 
claims 82 to 98, wherein one specificity thereof is for an agent effective to 
increase the half life of the ligand. 

100. A kit comprising a closed conformation multi-specific ligand according to 
any one of claims 82 to 99. 

101 . Nucleic acid encoding at least a closed conformation multispecific ligand 
according to any one of claims 82 to 99. 

1 02. A vector comprising nucleic acid according to claim 101. 

103. A vector according to claim 102, further comprising components 
necessary for the expression of a closed conformation multispecific ligand. 

1 04. A host cell transfected with a vector according to claim 1 03. 

105. A method for detecting the presence of a target molecule, comprising; 

(a) providing a closed conformation multispecific ligand bound to an agent, said 
ligand being specific for the target molecule and the agent, wherein the agent 
which is bound by the ligand leads to the generation of a detectable signal on 
displacement from the ligand; 

(b) exposing the closed conformation multispecific ligand to the target molecule; 
and 

(c) detecting the signal generated as a result of the displacement of the agent. 

106. A method according to claim 1 05, wherein the agent is an enzyme, which 
is inactive when bound by the closed conformation multispecific ligand. 

1 07. A method according to claim 1 05, wherein the agent is the substrate for an 
enzyme 

1 08. A method according to claim 1 07, wherein the agent is a fluorescent, 
luminescent or chromogenic molecule which is inactive or quenched when bound 
by the ligand. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



143 



PCT/GB2003/002804 



1 09. A kit for performing a method according to any one of claims 1 05-1 08, 
comprising a closed conformation multispecific ligand capable of binding to a 
target molecule, and optionally an agent and buffers suitable therefor. 

110. A homogenous immunoassay incorporating a method according to any 
one of claims 105-108. 

111. A ligand according to any one of claims 1 to 56 for use in therapy. 

112. A pharmaceutical composition comprising a ligand according to any one 
of claims 1 to 56, and a pharmaceutically acceptable eccipient, carrier or diluent. 

113. A method for preparing a chelating multimeric ligand comprising the steps 
of: 

(a) providing a vector comprising a nucleic acid sequence encoding a 
single binding domain specific for a first epitope on a target; 

(b) providing a vector encoding a repertoire comprising second binding 
domains specific for a second epitope on said target, which epitope can be the 
same or different to the first epitope, said second epitope being adjacent to said 
first epitope; and 

(c) expressing said first and second binding domains; and 

(d) isolating those combinations of first and second binding domains which 
combine together to produce a target-binding dimer. 

114. A method according to claim 113, wherein the first and second binding 
domains are associated covalently through a linker. 

115. A method according to claim 113, wherein the first and second binding 
domains are associated non-covalently. 

116. A method according to claim 113, wherein the first and second binding 
domains are associated through natural association of the domains. 

117. A method according to claim 1 1 6, wherein the binding domains comprise 
a Vh domain and a V K domain. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



1 717 



PCT/GB2003/002804 



CD 
u_ 



o CJ 

PO CO CD 

K < 

H 
Cm E-i 

O 
Eh O 



cj 



o cd 
cd 

Eh 
CO O 
Eh 

O 

CD 

< CJ 
CD 

Eh 
CJ CD 
Eh 

O 
CO CJ 
Eh 

O O 
CM Eh 

_ < 
PS CD 

cd 

hP EH 

CJ 

u 

CO CJ 
Eh 

CD 

a cd 
cd 

cd 
cd co 
cd 

Eh 
Or O 
O 

CD 
O < 
CJ 

. < 

> Eh 
CD 

CD 
hP Eh 
Eh 

O CJ 
rH CD CD 
33 CD 

< 

CD CD 
CD 

CD 
CD CD 
CD 

EH 
CO CJ 
Eh 

a 

CD 

CD 
hP Eh 
Eh 

CD 
hP Eh 
CJ 

a 

CJ 

CD 
> Eh 
CD 

CD 

rH H < 

tn CD 



CJ 
Eh 



CJ 
•55 



Eh CJ 



CO 



CD 



a 

CD 
EH 

CD 
ICDJ 



Eh 
CD CD 
CD 



CO 



m cd 



CM 

LO CO 



EH 
CD 

MTV 

Eh 

CD 
CD 



Eh 
M Eh 



O 
PS 



a 

CD 



CM 

PS 
Q 
CJ 



CO CJ 
Eh 

CJ 
> Eh 
CD 

CD 
IS CD 
Eh 

CD 

CD 

CD 

1-3 Eh 

a 

CD 
CD CD 

a 

CD 

CD 
CD CD 
CD 

CM CJ 
CJ 

O H 
^ < U 
CD 

CD 

o 

CJ 

ps cd 

CJ 

> Eh 
CD 

a 

IS CD 
Eh 



CJ 
CO CD 
< 

CD 
2 Eh 
< 

CJ 

< a 

CD 

Eh 

Eh 

CJ 
co CD 



r-H 

Q 

CJ 







o 






Q 










3 








CD 








<; 








CD 








a 








CJ 








CD 






PS 


<; 






CD 








< 










ro 






CD 


rH 


o 


hP 


Eh 


rH 






a 








CJ 




JH 


r r\ 










< 








a 




fu 






















O 


CM 




CD 


rH 


oo 




Eh 


rH 


pa 






PP 




O 










a 




o 




CD 




oo 


hP 


Eh 




Pel 




O 








Eh 






>H 


< 








Eh 





CD 
i-P Eh 
CJ 

CD 
Eh CJ 
f=C 

CJ 

CD 

U 
CO CJ 
Eh 



a 

Q < 
CD 

<< 

PS CD 

o a 

t CO CJ 
Cn Eh 



CJ 
Eh 

pC 

CJ 
CJ 

< 
cj 

Eh 

Eh 



CD 
PS CD 
O 

a 

CD CD 
CD 



CD 



CD 
> Eh 
CD 

CJ 
CO CJ 
Eh 

CJ 
Q Ft; 
CD 

O < 
vo < CJ 
ffi CD 



Eh 
CD CD 
CD 

CJ 
CD CD 
CD 

*S 
CD CD 
CD 

Eh 

CD CD 
CD 

a 

CO CD 
< 

CD 
CO CJ 



a 

> Eh 
CD 



H 
O 



rH Eh 



CJ 

a 



o 
o 



CJ 
> Eh 
CD 

CD 
hP Eh 
CJ 

CJ 
Eh CJ 

< 
«; 

CD CD 
CD 

CD 

oj <: 
o 

CJ 
CD CD 
CD 

CD 
IS CD 
Eh 



CJ 

Eh 

CJ 
Q < 
CD 



Eh 
Eh 
Eh 



CO 



CD 



FT 

CJ 
CD 

Eh 
CD 
CD 

EH 

EH 

CD 



CO 



CD 
< CJ 
CD 

Eh 
CJ CD 
Eh 

CJ 

Eh 

o eh 

CTl >H < 

PC! Eh 

^ *C 
> Eh 
CD 

a 
*c a 

CD 

a 
eh a 



CO 

PS 

a 

CJ 

cn 



PS CD 
CJ 

a < 

CD 

CD CD 
CD 

^ < 
> Eh 

CD 

Eh 

CO CJ 

EH 

< CJ 
CD 

Eh 
CO CJ 
Eh 

CD 
hP Eh 
CJ 

O CJ 
rH CO CJ 
hP Eh 

CJ 
CO CJ 
Eh 

rtj 
P-i CJ 

a 

Eh 
CO u 
Eh 

CD 

CJ 

CJ 

eh a 
< 

CD 
S Eh 
< 

CD 
O < 

o 
a 

1 — I Eh 

o 

rH Q f< 

t-P CD 



CD 
CJ 



i — i 



co 



CD 



CD 
CD CD 
CD 

CJ 
CD CD 
CD 

Eh 
CD CD 
CD 

O 
CD CD 
CD 

O 
co CD 

o 

CD CD 
CD 

Eh 

CD CD 
CD 

CD CD 
CD 

CJ 
CD CD 
CD 

*C 
CO CJ 
Eh 



M 
i — I 

CO 



CJ 
Eh 

< 

CD 
Eh 
CJ 

O 
Eh 
CJ 

CD 



Eh 
P-i CJ 
CJ 

CJ 
< CJ 
CD 



CD 
CD CD 
CD 

O f< 
^ CM U 
HP CJ 



CD 

CJ 

CD 

CJ 

Eh 

Eh 

CD 
IS CD 
Eh 

Eh 

< 
hP Eh 
Eh 



Eh 

CJ 
CO CD 

O CJ 
ro co CD 
hP < 



rH 



EH 
EH 



a 

co CD 

< 

CD 

o 

Eh 

CO CD 

CD 

CD 
PS CD 
CJ 

CJ 
CJ CD 
Eh 

Eh 

o 

M Eh 

O CJ 
CNJ Eh CJ 
hp <{ 

CJ 

> EH 

CD 



rH 
P£ 
Q 
CJ 
hP 

















CD 


o 






• ■ 




rH 

1 1 


rv* 
Mh 






CJ 


hP 






Eh 








co 


CD 




S> 
hH 






< 








CJ 








co 


CD 




1 — | 

1 1 






< 










CJ 








M 


Eh 




rH 






< 








CJ 








Eh 


CJ 




> 






Ft: 










o 








hP 


Eh 










CJ 








Eh 








Eh 


CJ 




Fh 






< 








Ua 


CJ 








Eh 




CD 






Eh 












o 




o 




EH 


o 




r- 


Q 




rH 


N 


hP 




CD 


hP 






< 








Eh 


r ) 

V-/ 


















CD 








CD 


a 










CD 










Eh 








CO 


CJ 




Eh 






Eh 








< 








CD 


CD 










CD 







Eh 
CO CD 
«=£ 

O 
CD CD 
CD 

Eh 

co CD 
< 

CJ 
Ua Eh 
Eh 

PS CD 
< 

O FtJ 
VD CO CJ 
hP EH 

< 

CU CJ 
CJ 

CJ 
> Eh 

CD 

CD 
CD CD 
CD 



Eh 
CO CD 

cy Ft: 

CJ 

CD 
hP Eh 
Eh 



CO 



CD 



CJ 
co CJ 
Eh 

< CJ 
CD 



O 

to Ft: 
hP 



Eh 
cj 
£2 



CM 
PS 

Q 
CJ 
hP 



CD 
CD 
O 

< 



CJ 
Eh 
< 



CD 

CD 
Eh 
CD 

CD 



CJ 
CJ 
< 

CD 
CD 
CD 



O 

CJ 
CD 
CD 

CJ 



CD 
CJ 
< 



CM O 
CJ 



Eh 



CO 



CO 



o 

EH 

CD 
CJ 

CD 



o CD 
O < 
hP CJ 

o Ft; 

CJ 

Eh 
O CD 
Eh 

CJ 

>h Ft; 

Eh 

a 

EH 

Eh 
Eh CJ 

^ < 
< CJ 
CD 

Eh 
Cx4 Eh 
Eh 

Eh 
Q Ft; 
CD 



ro 

PS 
Q 
CJ 
hP 



CD 



Eh 
>n Pt; 
Eh 



O Eh 
CO CM O 
hP O 

< 

o < 
a 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



2/17 



PCT/GB2003/002804 



FIG. 2 



o 
o 



r 




lac RBS 
promoter 



leader 



linker peptide 
tag 



plT1/plT2 



■(colEI orj)- 



amp 



gin 



{M13ori> 



RBS 

C AG G AAAC AG C T AT G AC CAT GAT T AC G C C AAG CT T G CAT G C AAAT T C TAT T T C AAG GAG AC AGT CAT A ATG AAA TAC CTA 

> M K Y L 

LMB3 



Sfil 



Ncol 



TTG CCT ACG GCA GCC GCT GGA TTG TTA TTA CTC GC G GCC CAG CCG GCC ATG GCC GAG GTG TTT 
L PTAAAGLLLLAAQPAMAEVF 

Xhol linker 
GAC TAC TGG GGC CAG GGA ACC CTG GTC ACC GT C TCG AG C GGT GGA GGC GGT TCA GGC GGA GGT 
DYWGQGTLVTVS SGGGGSGGG 



Sail NotI 
GGC AGC GGC GGT GGC GG G TCG AC G GAC ATC CAG ATG ACC CAG GCG GCC GCA GAA CAA AAA CTC 

GSGGGGST DIQMTQAAAEQKL 

< ■ 

link seq new 

HIS -tag 

CAT CAT CAT CAC CAT CAC GGG GCC GCA 
HHHHHHGAA 
(insertion in pIT2 only) 

myc-tag Gene III 

ATC TCA GAA GAG GAT CTG AAT GGG GCC GCA TAG ACT GTT GAA AGT TGT TTA GCA AAA CCT CAT 

ISEEDLNGAA* TVESCLAKPH 

< 

pHEN seq 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



3/17 



PCT/GB2003/002804 



ro 

CM 
t-H 
CD 

CO 

[ — - 

LO 

co 

CN) 



co 



ON 



oo 



VJD 



CNJ 



on 

CO 

r— 

co 
O 

ro 

CNJ 

i-H 
CD 
ON 
CO 
C — 

LO 

oo 
csi 
t—t 

CD 

on 
co 
r— 



CNJ 

I 



CO 

■ 

(3 

Ll_ 



LO 



CNJ 



8 



CO 



CNJ 



ON 
OO 

r— 

VD 
LO 

co 

CNJ 
t-t 

CD 

on 

CO 

r— 



LO 

CO 
CnJ 



on 
oo 

VJD 
LO 
-TP 
CO 
CNJ 



on 
oo 

r- 
vr> 

LO 

CO 
CNJ 

! I 

CD 

cn 
oo 
r— 

LX> 

LO 

co 

CnJ 



CO 
CO 

£> 
E-h 

E-H 

cd 
oj 
cd 



pC 
CD 

>H 

CO 



s 

g 

E-h 

Q 

S 

CO 
£2 

s, 
oj 

1-3 

E-h 

s: 
co 
o 

D4 
CO 
! — I 
£~t 

on 



1 




1 
1 


i 


1 


i 


i 


1 


I 


t 

* 


1 


1 
1 


t 


1 


i 


■ 


j 


i 


t 


1 


j 


j 


J 


j 


J 


1 


1 


1 




I 


i 


1 




1 


1 


l 


1 




1 


i 


1 


i 


1 


1 


1 


1 




1 


i 


1 


t 


t 




J 


1 




I 


i 


1 


i 


» 


1 


1 


1 




I 


i 


I 


i 


! 


1 


r 


1 




I 


i 


1 


i 


1 


1 


j 


t 




1 


i 


J 




1 


1 


i 


1 




I 


i 


1 


i 


1 


1 


i 


1 




I 


i 


1 


i 


I 


J 


i 


1 




! 


i 


t 


i 


1 


1 




1 




J 


i 


1 


i 


1 


1 


l 




E — ' 


O-i 


<LD 


t> 


oj 


O 




co 




t-=\ 


pC 


i-3 


>-h* 


»-3 


k3 






i-=\ 


g 


cd 


cd 


:> 


:> 


k3 


I" "t 




cd 




an 


O-i 






CO 


15*5 





LO 
















-ecr* 
















CO 


> 














Cn] 


CO 
















Q 














CD 


SS 














ON 
















oo 




on 


CD 


CD 


os 


OS 


oj 


r— 


E-h 














vo 


CO 






pC 


an 






LO 


cd 


pC 


CO 






X 


CO 


^p 


cd 














CO 


CO 


>H 


E-h 


O-i 


CO 


E-h 




co 


cd 


cu 


PC 


CD 




04 


pC 


CNJ 


CO 


CO 


cd 




Cu 


DJ 


CO 


t-H 


l-Ht 


1 


i 


J 


1 


1 


1 


CD 


pc 


an 


Q 


on 


CO 







On 
I 



CO on 



PC 
CO 



T 



CO 

0 



> 

CO 
CO 
I 

E-h 



CO 

i-3 
CD 

cd 

D-i 

pC 

o 



CO 



CO 



CO 

E-« 

CD 
CO 

c_> 

CO 
h3 

o^ 

h3 
CO 
CD 
CD 
On 

> 

CD 
CD 
CD 
CO 

Da 

> 



CO 



CO 



CO 

e 



Cnj 

I 



CNJ 



' — i 



CO 

r— 

tO 

co 

CNJ 
l — I 
CD 
ON 
CO 



ON 



OO 



LO 



CO 



CNJ 



oo 
r- 
vo 

LO 

ro 

CN) 
r — H 
CD 
ON 
CO 
[ — 
NO 
LO 

co 

CNJ 

i — i 

CD 
ON 
CO 

r— 

LO 

CO 
CnI 
t— I 
CD 
ON 
OO 



VO 
LO 

oo 

CN) 



ON 
OO 

r— 

LO 

PO 
CNJ 



cn 
co 
r— 

VD 
tO 



co 

CNJ 

CD 
ON 
OO 

vo 

LO 



co 

CN) 

« — i 

CD 
ON 
CO 

r~- 

NO 
LO 

oo 

CN) 

CD 
ON 

OO 

r~- 

kO 

LO 

CO 
CN) 



i — i 

E-h 

CD 
OJ 
CD 





E-H 


1 


1 


1 


vo 




CD 






LO 


o_. 


) 


1 


i 




E-h 


CO 




Q 


ro 


CO 


cr; 




>-» 


CNJ 








^> 


i — i 


CO 


O-i 




on 


CD 










CTi 


ex 









>H 

CM 
O 

co 

CO 
t — I 

>-3 
E-i 

Q 
E-h 
CD 
CO 
CD 
CO 
CD 
CO 

on 

CO 

o-» 

CD 



CO 
O 
i-3 

co an 
co 



s 



ben 

O-i 



CD 
OJ 

o 



»-3 

CO 
CO 
}—i 
CO 
OJ 
CO 



o 

E-h 
>-H 
E-h 

CO 

f=C 

CO 

E3i 

CO 
CU 
CO 

OJ 
E-h 

OJ 

I — I 
Q 



on 



on h3 

h3 h< 



CO 

cn 

-i-H 
rd 

JG 

o 
an 




CO 









-a 








X3 


-.-I 








to 






-a 


CO 


fCJ 






t— i 


CD 


CO 


CO 


CO 


t-H 


-c 






i-H 






LO 


LO 


T— ) 


o 


CNJ 




O 


pC 


pC 


pC 






an 


an 


an 


an 


an 


an 


an 


an 




> 


:> 


> 


> 


> 




^> 


> 





3 

to 

OO LO oo 

> 5^ w o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



4/17 



PCT/GB2003/002804 



o 



1.6 
1.4 
1.2 
1 

0.8 
0.6 
0.4 
0.2 
0 



2*2*2*2*2 



1 



FIG. 4 

Phage ELISA of a dual specific ScFv antibody K8 



••2*2»2*2 

i*2*2*2*2 
»z*2*2*2* 



••J:-:*:* 

•I*I*2*2* 



4 5 
Antigens 



1- HSA 

2- APS 

3- b-gal 

4- Peanut 

5- BSA 

6- lysosyme 

7- cytochrome c 



FIG. 5 

Soluble ELISA of the Dual Specific ScFv Antibody K8 




0 100 200 300 400 500 600 



K8 ScFv concentration (nmol) 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



5/17 



PCT/GB2003/002804 



Q 
O 



1.6 
1.4 
1.2 

1 - 
0.8- 
0.6 
0.4- 
0.2- 

0 



FIG. 6 

Soluble ScFv ELISA of K8V K /dummy V H clone 



1 



-:!:!:-:3:s:2:J;S:ir: 



m m m m m m m 



2 3 
Antigens 



4 



1- BSA 

2- b-gal 

3- APS 

4- Protein A 



FIG. 7 



RBS 

CAGGAAACAGCTATGACCATGATTACGCCAAGCTTGCATGCAAATTCTATTTCAAGGAGACAGTCATA ATG AAA TAG CTA 
> M K Y L 

LMB3 

Sfil Ncol 



TTG CCT ACG GCA GCC GCT GGA TTG TTA TTA CTC GC G GCC CAG CCG GCC ATG GCC GAG GTG TTT 
LPTAAAGLLLLA A ~ O P A M A E V F 



Xhol linker 
GAG TAC TGG GGC CAG GGA ACC CTG GTC ACC GT C TCG AG C GGT GGA GGC GGT TCA GGC GGA GGT 
DYWGQGTLVTVS SGGGGSGGG 



Sail NotI 
GGC AGC GGC GGT GGC GG G TCG AC G GAC ATC CAG ATG ACC CAG GCG GCC GCA GAA CAA AAA CTC ' 
GSGGGGSTT) I QMTQAAAEQKL 

< 

link seq new 

HIS -tag 

CAT CAT CAT CAC CAT CAC GGG GCC GCA 
HHHHHHGAA 
(insertion in V domain vector 2 only 



myc-tag Gene III 

ATC TCA GAA GAG GAT CTG AAT GGG GCC GCA TAG ACT GTT GAA AGT TGT TTA GCA AAA CCT CAT 
ISEEDLNGAA * TVESCLAKPH 

< 

pHEN seq 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



6/17 



FIG. 8 



PCT/GB2003/002804 



lac RBS 
promoter 



■Q p15 ori J: 



8 




leader linker 



CH vector 



CH gene 



{M13 ori) - 



o 

CD 
CO 



— Q: 

0)32 

QQ 5 Uj 




myc 2x 
tag ochre 



Cm 



FIG. 9 



r 



o 
o 



CD 

CO CO 




lac RBS 
promoter 



leader 



CK vector 



■ (colEI orj) - 



amp 



* ■ * p 



CK gene 



CD 
O 

CO 



o 



flag 
tag 



2x 

ochre 



<M13 ori> 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



7/17 



PCT/GB2003/002804 



o 
o 



O) 



FIG. 10 

TNF receptor assay 




0.1 1 10 100 1000 

dAb/dimer concentration nM 



PEP1-5d4 
PEP1-5-19d4 
PEP1-519 monomer 



o 120 



o 
o 



FIG. 11 

TNF receptor assay 




1 10 100 

dimer concentration nM 



1000 



PEP1-5d1 
PEP1-5d2 
PEP1-5d3 
PEP1-5d4 
PEP1-5d5 
PEP1-5d6 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



8/17 



PCT/GB2003/002804 



FIG. 12 



110 



o 
o 



-10 



TNF Receptor assay 




*-PEP1-5-19 3U homodimer 
PEP1-5-19 5U homodimer 
PEP1-5-19 71) homodimer 



*-PEP1-5-19 CH/CK homodimer 
h*-PEP1-5-19 cys hinge 



0.1 1 10 100 1000 

dAb/dimer concentration nM 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

9/17 



FIG. 13 



Dummy V H sequence for library 1 



1 


E 
GAG 
CTC 


V 
GTG 
CAC 


Q 
CAG 
GTC 


L 
CTG 
GAC 


L 
TTG 
AAC 


E 

GAG 
CTC 


S 
TCT 
AGA 


G 
GGG 
CCC 


G 
GGA 
CCT 


G 
GGC 
CCG 


L 
TTG 
AAC 


V 
GTA 
CAT 


Q 
CAG 
GTC 


P 
CCT 
GGA 


G 
GGG 
CCC 


G 
GGG 
CCC 


49 


S 
TCC 
AGG 


L 
CTG 
GAC 


R 
CGT 
GCA 


L 
CTC 
GAG 


S 
TCC 
AGG 


C 
TGT 
ACA 


A 
GCA 
CGT 


A 
GCC 
CGG 


S 
TCC 
AGG 


G 
GGA 
CCT 


F 
TTC 
AAG 


T 
ACC 
TGG 


F 
TTT 
AAA 


S 
AGC 
TCG 


s 

AGC 
TCG 


Y 
TAT 
ATA 


97 


A 
GCC 
CGG 


M 
ATG 
TAC 


S 
AGC 
TCG 


W 
TGG 
ACC 


V 
GTC 
CAG 


R 
CGC 
GCG 


Q 
CAG 

GTC 


A 
GCT 
CGA 


P 
CCA 
GGT 


G 
GGG 
CCC 


K 
AAG 
TTC 


G 
GGT 
CCA 


L 
CTA 
GAT 


E 
GAG 
CTC 


W 
TGG 
ACC 


V 
GTC 
CAG 




S 


A 


I 


S 


G 


S 


G 


G 


S 


T 


Y 


Y 


A 


D 


S 


V 


145 


TCA 


GCT 


ATT 


AGT 


GGT 


AGT 


GGT 


GGT 


AGC 


ACA 


TAC 


TAC 


GCA 


GAC 


TCC 


GTG 




AGT 


CGA 


TAA 


TCA 


CCA 


TCA 


CCA 


CCA 


TCG 


TGT 


ATG 


ATG 


CGT 


CTG 


AGG 


CAC 


193 


K 
AAG 
TTC 


G 
GGC 
CCG 


R 
CGG 
GCC 


F 
TTC 
AAG 


T 
ACC 
TGG 


I 

ATC 
TAG 


S 
TCC 
AGG 


R 
CGT 
GCA 


D 

GAC 
CTG 


N 
AAT 
TTA 


S 
TCC 
AGG 


K 
AAG 
TTC 


N 
AAC 
TTG 


T 
ACG 
TGC 


L 
CTG 
GAC 


Y 
TAT 

jAT.A 


241 


L 

CTG 
GAC 


Q 

CAA 
GTT 


M 
ATG 
TAC 


N 
AAC 
TTG 


S 

AGC 
TCG 


L 
CTG 
GAC 


R 
CGT 
GCA 


A 
GCC 
CGG 


E 
GAG 
CTC 


D 
GAC 
CTG 


T 
ACC 
TGG 


A 
GCG 
CGC 


V 
GTA 
CAT 


Y 
TAT 
ATA 


Y 
TAC 
ATG 


c 

TGT 
ACA 




A 


K 


S 


Y 


G 


A 


F 


D 


y 


W 


G 


Q 


G 


T 


L 


V 


289 


GCG 


AAA 


AGT 


TAT 


GGT 


GCT 


TTT 


GAC 


TAC 


TGG 


GGC 


CAG 


GGA 


ACC 


CTG 


GTC 




CGC 


TTT 


TCA 


ATA 


CCA 


CGA 


AAA 


CTG 


ATG 


ACC 


CCG 


GTC 


CCT 


TGG 


GAC 


CAG 


337 


T 
ACC 
TGG 


V 
GTC 
CAG 


S 
TCG 
AGC 


S 
AGC 
TCG 



























SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



10/17 



PCT/GB2003/002804 



FIG. 14 



Dummy V H sequence for library 2 



1 


E 
GAG 
CTC 


V 
GTG 
CAC 


Q 
CAG 
GTC 


L 
CTG 
GAC 


L 
TTG 
AAC 


E 
GAG 
CTC 


S 
TCT 
AGA 


G 
GGG 
CCC 


G 
GGA 
CCT 


G 
GGC 
CCG 


L 
TTG 
AAC 


V 
GTA 
CAT 


Q 
CAG 
GTC 


P 
CCT 
GGA 


G 
GGG 
CCC 


G 
GGG 
CCC 


49 


S 
TCC 
AGG 


L 
CTG 
GAC 


R 
CGT 
GCA 


L 
CTC 
GAG 


S 
TCC 
AGG 


C 
TGT 
ACA 


A 
GCA 
CGT 


A 
GCC 
CGG 


S 
TCC 
AGG 


G 
GGA 
CCT 


F 
TTC 
AAG 


T 
ACC 
TGG 


F 
TTT 

.TVAAl 


S 
AGC 
TCG 


S 
AGC 
TCG 


Y 
TAT 
ATA 


97 


A 
GCC 
CGG 


M 
ATG 
TAC 


S 
AGC 
TCG 


W 
TGG 
ACC 


V 
GTC 
CAG 


R 
CGC 
GCG 


Q 
CAG 
GTC 


A 
GCT 
CGA 


P . 
CCA 
GGT 


G 
GGG 
CCC 


K 
AAG 
TTC 


G 
GGT 
CCA 


L 
CTA 
GAT 


E 
GAG 
CTC 


W 
TGG 
ACC 


V 
GTC 
CAG 




S 


A 


I 


S 


G 


S 


G 


G 


S 


T 


Y 


Y 


A 


D 


S 


V 


145 


TCA 


GCT 


ATT 


AGT 


GGT 


AGT 


GGT 


GGT 


AGC 


ACA 


TAC 


TAC 


GCA 


GAC 


TCC 


GTG 




AGT 


CGA 


TAA 


TCA 


CCA 


TCA 


CCA 


CCA 


TCG 


TGT 


ATG 


ATG 


CGT 


CTG 


AGG 


CAC 


193 


K 
AAG 
TTC 


G 
GGC 
CCG 


R 
CGG 
GCC 


F 
TTC 
AAG 


T 

ACC 
TGG 


I 

ATC 
TAG 


S 
TCC 
AGG 


R 
CGT 
GCA 


D 
GAC 
CTG 


N 
AAT 
TTA 


S 
TCC 
AGG 


K 
AAG 
TTC 


N 
AAC 
TTG 


T 
ACG 
TGC 


L 

CTG 
GAC 


Y 
TAT 
ATA 


241 


L 
CTG 
GAC 


Q 
CAA 
GTT 


M 
ATG 
TAC 


N 
AAC 
TTG 


S 
AGC 
TCG 


L 
CTG 
GAC 


R 
CGT 
GCA 


A 
GCC 
CGG 


E 
GAG 
CTC 


D 
GAC 
CTG 


T 
ACC 
TGG 


A 
GCG 
CGC 


V 
GTA 
CAT 


Y 
TAT 

A.TAi 


Y 
TAC 
ATG 


C 
TGT 
ACA 




A 


K 


S 


y 


G 


A 


X 


X 


X 


X 


F 


D 


Y 


W 


G 


Q 


289 


GCG 


AAA 


AGT 


TAT 


GGT 


GCT 


NNK 


NNK 


NNK 


NNK 


TTT 


GAC 


TAC 


TGG 


GGC 


CAG 




CGC 


TTT 


TCA 


ATA 


CCA 


CGA 


NNK 


NNK 


NNK 


NNK 


AAA 


CTG 


ATG 


ACC 


CCG 


GTC 


337 


• 

G 
GGA 
CCT 


T 
ACC 
TGG 


L 
CTG 
GAC 


V 
GTC 
CAG 


T 

ACC 
TGG 


V 
GTC 
CAG 


S 
TCG 
AGC 


S 
AGC 
TCG 



















SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



11 / 17 



PCT/GB2003/002804 



FIG. 15 



Dummy V K sequence for library 3 



1 


D 
GAC 
CTG 


I 

ATC 
TAG 


Q 
CAG 
GTC 


M 
ATG 
TAC 


T 
ACC 
TGG 


Q 
CAG 

GTC 


S 
TCT 
AGA 


P 
CCA 
GGT 


S 
TCC 
AGG 


S 
TCC 
AGG 


L 
CTG 
GAC 


S 
TCT 
AGA 


A 
GCA 
CGT 


S 
TCT 
AGA 


V 
GTA 
CAT 


G 
GGA 
CCT 




D 


R 


V 


T 


I 


T 


C 


R 


A 


S 


Q 


S 


I 


S 


S 


Y 


49 


GAC 


CGT 


GTC 


ACC 


ATC 


ACT 


TGC 


CGG 


GCA 


AGT 


CAG 


AGC 


ATT 


AGC 


AGC 


TAT 




CTG 


GCA 


CAG 


TGG 


TAG 


TGA 


ACG 


GCC 


CGT 


TCA 


GTC 


TCG 


TAA 


TCG 


TCG 


ATA 


97 


L 

TTA 
AAT 


N 
AAT 
TTA 


W 
TGG 
ACC 


Y 
TAC 
ATG 


Q 
CAG 
GTC 


Q 
CAG 
GTC 


K 
.AA.A 
TTT 


P 
CCA 
GGT 


G 
GGG 
CCC 


K 
AAA 
TTT 


A 
GCC 
CGG 


P 
CCT 
GGA 


K 
AAG 
TTC 


L 

CTC 
GAG 


L 

CTG 
GAC 


I 

ATC 
TAG 


145 


Y 
TAT 


A 
GCT 


A 
GCA 


S 
TCC 


S 
AGT 


L 
TTG 


Q 
CAA 


S 
AGT 


G 
GGG 


V 
GTC 


P 
CCA 


S 
TCA 


R 
CGT 


F 
TTC 


S 
AGT 


G 
GGC 




ATA 


CGA 


CGT 


AGG 


TCA 


AAC 


GTT 


TCA 


CCC 


CAG 


GGT 


AGT 


GCA 


AAG 


TCA 


CCG 


193 


S 
AGT 
TCA 


G 
GGA 
CCT 


S 
TCT 
AGA 


G 
GGG 

ccc 


T 
ACA 
TGT 


D 
GAT 
CTA 


F 
TTC 
AAG 


T 
ACT 
TGA 


L 
CTC 
GAG 


T 
ACC 
TGG 


I 

ATC 
TAG 


S 
AGC 
TCG 


S 
AGT 
TCA 


L 
CTG 
GAC 


Q 
CAA 

GTT 


P 
CCT 
GGA 




E 


D 


F 


A 


T 


Y 


Y 


C 


Q 


Q 


S 


Y 


S 


T 


P 


N 


241 


GAA 


GAT 


TTT 


GCT 


ACG 


TAC 


TAC 


TGT 


CAA 


CAG 


AGT 


TAC 


AGT 


ACC 


CCT 


AAT 




CTT 


CTA 


AAA 


CGA 


TGC 


ATG 


ATG 


ACA 


GTT 


GTC 


TCA 


ATG 


TCA 


TGG 


GGA 


TTA 


289 


T 
ACG 
TGC 


F 
TTC 

AAG 


G 
GGC 
CCG 


Q 
CAA 
GTT 


G 
GGG 
CCC 


T 

ACC 
TGG 


K 
AAG 
TTC 


V 
GTG 
CAC 


E 
GAA 
CTT 


I 

ATC 
TAG 


K 
AAA 
TTT 


R 
CGG 
GCC 











SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



12/17 



PCT/GB2003/002804 



FIG. 16 

Nucleotide and amino acid sequence of anti MSA dAbs MSA 16 and MSA 26 

A: MSA 16 



GAC 
D 


ATC 
I 


CAG 
Q 


ATG 
M 


ACC 
T 


CAG 
Q 


TCT 
S 


CCA 
P 


TCC 
S 


TCC 
S 


CTG 
L 


TCT 
S 


GCA 
A 


TCT 
S 


GTA 
V 


GGA 
G 


GAC 
D 


CGT 
R 


GTC 
V 


ACC 
T 


ATC 
I 


ACT 
T 


TGC 
C 


CGG 
R 


GCA 
A 


AGT 
S 


CAG 

Q 


AGC 
S 


ATT 
I 


ATT 
I 


AAG 
K 


CAT 
H 


TTA 
L 


AAG 
K 


TGG 

w . 


TAC 
Y 


CAG 
Q 


CAG 

Q 


AAA 
K 


CCA 
P 


GGG 
G 


AAA 
K 


GCC 
A 


CCT 
P 


AAG 
K 


CTC 
L 


CTG 
L 


ATC 
I 


TAT 
Y 


GGT 
G 


GCA 
A 


TCC 
S 


CGG 
R 


TTG 
L 


CAA 
Q 


AGT 
S 


GGG 
G 


GTC 
V 


CCA 
P 


TCA 
S 


CGT 
R 


TTC 
F 


AGT 
S 


GGC 
G 


AGT 
S 


GGA 
G 


TCT 
S 


G 


ACA 
T 


GAT 
D 


TTC 
F 


ACT 
T 


CTC 
L 


ACC 
T 


ATC 
I 


AGC 
S 


AGT 
S 


CTG 
L 


CAA 
Q 


CCT 
P 


GAA 
E 


GAT 
D 


TTT 
F 


GCT 
A 


ACG 
T 


TAC 
Y 


TAC 
Y 


TGT 
C 


CAA 

Q 


CAG 
Q 


GGG 
G 


GCT 
A 


CGG 
R 


TGG 
W 


CCT 
P 


X~ "< "7\ ✓^l 

CAG 

Q 


ACG 
T 


TTC 
F 


GGC 
G 


CAA 

Q 


GGG 
G 


ACC 
T 


AAG 
K 


f*n /""l 

GTG 
V 


GAA 
E 


ATC 
I 


AAA 
K 


CGG 
R 










B: MSA 26 
























GAC 
D 


ATC 
I 


CAG 
Q 


ATG 
M 


ACC 
T 


CAG 

Q 


TCT 
S 


CCA 
P 


TCC 
S 


TCC 
S 


CTG 
L 


TCT 
S 


GCA 
A 


TCT 
S 


GTA 
V 


GGA 
G 


GAC 
D 


CGT 
R 


GTC 
V 


ACC 
T 


ATC 
I 


ACT 
T 


TGC 
C 


CGG 
R 


GCA 
A 


AGT 
S 


CAG 

Q 


AGC 
S 


ATT 
I 


TAT 
Y 


TAT 
Y 


CAT 
H 


TTA 
L 


AAG 
K 


TGG 
W 


TAC 
Y 


CAG 

Q 


CAG 
Q 


AAA 
K 


CCA 
P 


GGG 
G 


AAA 
K 


GCC 
A 


CCT 
P 


AAG 
K 


CTC 
L 


CTG 
L 


ATC 
I 


TAT 
Y 


AAG 
K 


GCA 
A 


TCC 
S 


ACG 
T 


TTG 
L 


CAA 

Q 


AGT 
S 


GGG 
G 


GTC 
V 


CCA 
P 


TCA 
S 


CGT 
R 


TTC 
F 


AGT 
S 


GGC 
G 


AGT 
S 


GGA 
G 


TCT 
S 


GGG 
G 


ACA 
T 


GAT 
D 


TTC 
F 


ACT 
T 


CTC 
L 


ACC 
T 


ATC 
I 


AGC 
S 


AGT 
S 


CTG 
L 


CAA 

Q 


CCT 
P 


GAA 
E 


GAT 
D 


TTT 
F 


GCT 
A 


ACG 
T 


TAC 
Y 


TAC 
Y 


TGT 
C 


CAA 
Q 


CAG 
Q 


GTT 
V 


CGG 
R 


AAG 
K 


GTG 
V 


CCT 
P 


CGG 
R 


ACG 
T 


TTC 
F 


GGC 
G 


CAA 
Q 


GGG 
G 


ACC 
T 


AAG 
K 


GTG 
V 


GAA 
E 


ATC 
I 


AAA 
K 


CGG 
R 











SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



13/17 



PCT/GB2003/002804 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



14/17 



PCT/GB2003/002804 



o 

o 



CO 
CM 

< 

CO 



o 

s 

o 
o 

05 



CD I 



o 

_Q 









CM 


CO 


CD 


o> 


v ~ 


1 


i 


i 


i 


CO 
1 


CO 


CO 


CO 




I 


1 


1 


II 


ii 


II 


ii 


o 






\ > 


Lo- 


LL 


LL 


LL 








• m, mm 


ci 






c 


CO 


CD 


CO 


CO 


CM 


CM 


CM 


CM 


<D 


0 


CD 


CD 








c 


o 


O 


o 


o 


o 


O 


o 


o 


CO 


CO 


CO 


CO 


o 

• 


O 

• 


o 

■ 


o 

• 










■ 


• 


• 


■ 

1^. 



CO 
I 

XT 
II 

o 



CD 
CM 



_o 

O 
CO 

o 



i 

CO 
I 

II 

o 



CO 
CM 



CD CD 



O 
O 

CO 
O 



CO 
i 

II 
O 
LL 



CO 
CM 

CD 
C 

_o 

o 

CO 











CM 


LO 


CO 




i 


■ 


i 


i 


CO 


CO 


CO 


CO 


t 


1 


1 


1 


M 








II 


n 


ii 


II 




O 


o 


o 


LL 


LL 


10- 


LL 


m ■ ■■ 














c 


CD 


CO 


CO 


CO 


CM 


CM 


CM 


CM 


CD 


CD 


CD 


CD 










O 


O 


O 


O 


O 


O 


O 


O 


CO 


CO 


CO 


CO 


O 

■ 


O 

■ 


O 

■ 


o 

■ 










• 


• 


• 

1^- 


• 



CO 

II 

o 



CO 
CM 

0) 

c, 

o 
o 

CO 




o 

LO 
CO 



o 
o 

CO 



m 

CM 



CD 

E 



o 

CM 



-LO 



— > o 

LL. CM 



i — 1 — i — 1 — i — ■ — r 
o c 



o 

LO 



CM 



~i — 1 — r 

o c: 

LO 



LO 



Resp. Diff. 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



15/17 



PCT/GB2003/002804 




SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



16/17 



PCT/GB2003/002804 



(a) 
0.35 

0.3 
0.25 

0.2 
0.15 

0.1 
0.05 
0 



FIG. 19 



dSeriesI 



TAR 1 -5- TAR 1 -5- Dual 
1 9dAb 1 9dAb specific 
+ MSA 



Dual 
specific 
+ MSA 



(b) 



biotinylated anti-TNF 



TNF 




chromogenic 
substrate 



Streptavidin-HRP 



TNFRI/Fc chimera 



Anti-Fc capturing antibody 



TNF Receptor assay 
100t 




LH LH+ TNF + MSA 

MSA MSA only 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 PCT/GB2003/002804 

17/17 



FIG. 20 

TNF Receptor assay 
120 n 




Serum Albumin (mg/ml) 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



1/13 



PCT/GB2003/002804 



CD 

P 

CO 



LU 
LU 

cy 

LU 

CO 









s 


Ph 


rh 






CJ 






CJ 


cd 


CD 


rn 
CD 


CD 




CD 


fa fiC, 


4. j 


CD 


CD 














CD 


CD 




Eh 


> 




CJ 




CD 


03 CD 




CJ 


< 


CO 


CJ 






Eh 






CJ 




Q 




CD 




CD 


Pi CD 






r3 


CJ 


Eh 


CD 


fa Eh 




CJ 






< 


<; 




EH 


CD CD 




r i 
CJ 


CD 


?H 




CJ 












r ■ 

H 


tH 




r \ 
\J 










CJ 




CO 










rf; CJ 




tH 


a 


CD 


CD 






r n 

CD 


CJ CD 




CJ 


EH 


CO 


rn 

C9 


CJ 




s 


CO CJ 




EH 


H 




r \ 
CJ 




Pa 




u 




a 


A EH 




CJ 


CJ 


CD 


CD 


Eh 




r n 


Ph CD 






r \ 


pH 




rn 




Eh 


h-l EH 




Eh 


CJ 


H 




r \ 

CJ 




8 






U 


Eh 


CO 




CD 






CD CD 






CD 


CO 




CD 






CD CD 






CD 


> 


Eh 


Eh 




CD 


CJ 




CD 






CD 


r i 


is 


CD 




Eh 






o 


fa 




< 






> EH 






CD 




Eh 


CD 




CJ 


A EH 




Eh 


Eh 


CD 


CD 






CJ 


CD 






S3 


CD CD 


CD 


1 


CD 




CD 






CD CD 




8 








5 




CJ 


CO CJ 


< 


EH 


Eh 


CJ 




CD 


fa << 




CD 


CD 




CD 






rH* EH 


pJ 


a 


Eh 


CD 


CD 


CJ 


Eh 




CJ 








J o 


> 


EH 


CD 




CD 


o»g 




CD 




CD 


CD 




Eh 


> Eh 




CJ 


CD 


CD 


CD 


A CD 




CD 






CD 


ffl CD 


• 


EH 



a 

CO CD 

CD 
CO CJ 
EH 
CJ 
> Eh 

8 

EH CJ 



> EH 
CD 
_ CD 

r-H EH 

CJ 

cj 

CJ 

EH 



CD- CD 
CD 
CD 

a 



fa 



EH 

CD CD 
CD 

CD 
& CD 
Eh 
Eh 

EH 

Eh 
CJ 
CD CD 
CD 
CD 

a Eh 
CJ 
CJ 

fa CJ 
CJ 



CO 



CD 

CD 

CD 
CJ 



Eh 
Eh 
Eh 
CJ 
CD 
fa CJ 
CJ 
CD 

CD 



Eh 



Eh 



EH 

CJ 



fa 



H 

CD 
Eh 



a 

Eh CJ 

CJ 

fa EH 

a 

CD CD 

8 

CO CJ 
Eh 
CJ 
CJ 



9 

S 
CD 
Eh 
U CD 
Eh 
CJ 

co cj 
Eh 

CJ 
a Eh 
U 
H 
Pi CD 
CJ 
CD 
A H 
CJ 
CJ 

CO u 
Eh 
CD 

CD CD 
CD 



CD 



04 



CD 



Eh 
CJ 

CJ 
CD 



Eh 
CD 
CD 
Eh 
EH 



CJ 
CD CD 

fit 
CD CD 
CD 
CD 

u 8 

Eh 

CO CJ 
Eh 
CD 

fa rt! 
CD 
CD 

^ Eh 
Eh 



O 
CD 



in 
i 

EH 



Of 



CD 

> ^ 
CD 
CD 

CD 



* 8 



CD 



CJ 



CD 



01 S 
Eh 
CJ 

> ^ 
CD 

CD 

S CD 
Eh 
CD 

J Eh 
CJ 

CD 



CD 



CD 



1 
1 

CD 

1 

o 



, Eh 
rtj CJ 
CD 
CD 

a 



CJ 
CJ 





CD 




CD 




Eh 




> 










CD 


r< CJ 




CJ 




CD 


CO 


CJ 










U 










Eh 


Q 


1 






CD 




1 










CJ 






CD 


>* 












CJ 


















> 


Eh 




CD 




CD 


IS 


1 


< 


a 




CJ 






CD 


Eh 






CJ 






EH 












HH- 


B 










p 






CD 






PS 


CD 




CD 




m 






1 




CD 


CD 






CJ 






u 




g 




CD 








Eh 












Pi 


CD 




CD 




CJ 


a 






CD 


6 




H 








a 


CO 






CJ 






CO 


S3 










H 














< 




S5 






Eh 








EH 








E-! 


S 


EH 



Oi 



H 



H 



a 

CJ 
CD CD 
CD 



u 

Eh 
Eh 
CD 



E-« CJ 
f< 
Eh 

Pi CD 
U 
Eh 

CH O 
U 
CD 
Eh 



CJ 
CD 



Eh 
Eh 
Eh 

CD 



fa 



CD 
O fit 
CJ 

Of 
U 
Eh 

U CD 

Eh 

Eh 

CD 
Eh CJ 

o 



fa 



CD 
Eh 



Eh 
CD 



JxJ 



CD 



Eh 
04 CJ 
CJ 



CJ 
CD 
Eh 
O 
H 



CO CD 

co O 

CJ 
H Eh 

6 

CJ 



6 

Eh 
CJ 
Eh 
O 



fa 

Q 
EH 
CD 



EH 
EH 



pj CD 
U 

H Eh 



fa 



CD 

CD 
CD 

U 
Eh CJ 

CD 
CD CD 
CD 



lO 



o 

r4 



in 
cN 



o 

CO 



m 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



2/13 



PCT/GB2003/002804 



i 




H 

LO 



H 
O 



H 
LO 
t— I 



O 



to 







H 












H 








cd 














12! 










< 










CD 












o 




a 








u 




a 








CD 




CD 












CD 










CD 










CD 




CD 






a 


CJ 




O 




J CO 


3 


CD 


CD 


CD 


H 




< 


H 




CD 


tti 




CD 


O 




CD 




1 CO 


O 


U 


CD 


CD 






En 






CD 




O 






CD 




1 > 


Eh 






En 






CD 










• 


O 


H 




H 






H 


LO 




O 






LO 


CM 




ro 






m 




0 












CN 











& 



01 



a) 



to 



ro to 



CTi 
H 
I 

Lf) 
I 

H 




Oi 



co 



lh <: 

o a 

f=C Eh 

<C Eh 

CJ a 

CD CJ 

CD CJ 

CD CJ 



Cm 



CJ 
CD 
O Eh 
Eh 
CJ 

EH fit, 

CJ 
E-J 

cj 
u 

Eh < 
CJ 
Eh 

> CD 
Eh 
CD 

Pi CJ 

u 



CD CD 

CD 

CJ 

5 M 

CD 

Eh a 

a 

CD 
CD 
B 
CD 
< 
CJ W 

CD rfj 
CD 



< Eh 

Q CD CJ CO 

< Eh 
CD CJ 

CD CD CJ >n 

< H 

> CD S M 

Eh rf 

U CD 



W £h 
pa! 
CJ 
< CD 
Eh 



CO 



CJ 
Eh 
CD 
Eh 
CJ 

Eh 

a 

Eh 
CJ 
CJ 

s 

Cm CJ 
Eh 



CO 



CO 



Eh 

CD _ 

CJ HH 

CD 

u 
•3 

CD 

CM 

•=£ 
CD 

CD 
CD 

Eh 
CD 

CD CD 



CJ CD 

CO Eh < Aj 

CD CJ 

< Eh 

O* CJ CD W 

a cd 

CJ CD 

eh < eh a 

CD CJ 

Eh < 



S < Eh 
CD CJ 
f3 Eh 

a o cd 

U CD 
Eh 

Eh 
O CD 
Eh 
CD CJ 



a 



CO 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



3/13 



PCT/GB2003/002804 



u o 

H < 

ft H 

CD U 



fa 



ft 



cj o 
ft EH 



Eh < 

^ y P 

g g 

Eh CD O 



fa 



to- 



ft 



CO 



Pi 



ft Eh 

U CD 

CJ C3 

Eh rtJ 

CD U 

CD U 

Eh ft 

Eh g 

rtj Eh 



fa 



Q Eh 



9 






Eh 




a 


8 


Eh 








CD 


U 




u 


CD 


a 


CD 


CD 


U 


6 






U 






8 


CD 


Eh 


fa 


Eh 




8 




EH 








CD 





fa 



H 



fa 



U Eh 



ft 



U CD 



CO 



fa 



eh 

CD CJ 

CJ O 

CD U 

CD CJ 

Eh < 

CD CJ 

Eh ft 

CD CJ 

Eh ft 
Eh 

CD a 



a 



H 



CJ CD 
Eh 

rf! Eh 



CJ CD 

CO CJ CD 

g g 

*3 8 CD 



H 



CO 



ft 



co CJ CD 

eh ft 

CD U 

C <C Eh 



Eh CJ CD 
Eh 

CD CJ 
Eh ft 



ft 



CJ 



CD 



CD 

6 



JH EH 

Eh rfj 
CJ CD 

Eh < 

CD CJ 

Eh CJ CD 

< Eh 

8g 



fa 



CJ O 
CD CJ 



rfj Eh ft 
CD ft CJ CD 



CD co 

CD 

CD 

CJ CO 



CD CJ 

ft Eh 

CJ CD 

CD CJ 
Eh 

G CD 

< Eh 

CJ CD 

CJ CD 



Pi 



6 



CD 



U 

0) 
^! 
PI 

•ri 
i— 1 



a) 

-rl 
l-l 

to 



M 

■rl 
t-i 

m 



i 

I 

a 



fa 


















CD 


a 


CO 


a 


52 






Eh 










eh 


S 


OJ 








Eh 


1 












S 




Eh 




CD 


a 


a 




U 


CD 




CD 


.CJ 




CD 


u 


CD 


CD 


U 


< 


CJ 


CD 




Eh 


& 




CD 


U 




CD 




CO 




CJ 




CJ 














Eh 


fa 


Eh 


Q 


S 


Eh 




U 


CD 




Eh 






CD 


CJ 




CD 


CJ 




CD 






Eh CJ CD 

g g 

> Eh § 

CD CJ 

Eh f3 

ft CJ CD 

CJ CD 

eh 

CD CJ 



a i< Eh 

CJ CD 

Eh < 

CO CD CJ 

rtj Eh 

<C Eh 

rij O CD 

CD CJ 

CD CJ 

CD CJ 



ft Eh 

CO CJ CD 

Eh et 

ft H 

CJ CD 

U CD 

CJ CD 

Eh ft 

CD U 

CD CJ 



Pi 



a 



ft 



CJ CD 

CD U 

<! Eh 

CJ CD 

CD U 

Eh 

Eh < 

CD O 




CJ CD 

Eh CJ CD 

ft Eh 

CJ CD 

> Eh 

CD CJ 

Eh ft 

Pi CD CJ 

a cd 

a cd 

O ft Eh 

cd a 

< Eh 

CD CD CJ 

CD CJ 

ft Eh 

> Eh ft 
CD CJ 
Eh ft 
U CD 



CO 



Eh ft 

CD CJ 

p< cd a 

CJ CD 

CJ CD 

CO CJ CD 

EJ ft 

J f3 Eh 

f=3l CJ CD 

CD CJ 

EH ft 

Eh CJ CD 

Eh rtj 

CJ CD 

Eh < 

f< Eh 

CD U 



< Eh 
Eh pC 
CJ CD 

?h rtj &j 

EH l< 

cd a 

Eh CJ CD 

< Eh 
Eh ft 
CJ CD 



fa 



H 




Eh 

CD 



CD CJ 

Eh rtj 

CO CJ CD 

Eh fsC 

CD CJ 

h3 B ft 

CJ CD 

CJ CD 

CO CJ CD 

Eh ft 

CJ CD 

CO CJ CD 

Eh ft 

< Eh 
ft CJ CD 

CJ CD 

Eh < 

CO CJ CD 

Eh ft 

CD CJ 

a ft Eh 

O CD 

CJ CD 

Eh CJ CD 

< Eh 
CD CJ 

EH ft 

2 

ft, Eh 

CD CJ 

CJ CD 

EJ < 

ft, Eh 

CJ CD 

< Eh 
CD CJ 



Eh 
CJ 
CJ 

B 

CD 



Eh rfj 
CD ft CJ CD 



CD CJ 
CD CJ 
CJ CD 



W 




CD CJ 

CD CJ 

' Eh 
Eh 

CJ CD 

CJ CD 

ft Eh 

cd a 

CD CJ 

CD CJ 




CJ CD 

CJ CD 

CJ CD CJ 

Eh ft 

Eh ft 

CJ CD 

ft Eh 

CJ CD 

Eh rf 

rij Eh 



Eh 

CO CJ CD 

a .„ 

ft CJ CD 

U CD 

U CD 

Eh ft 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh ft 

CO CD CJ 



H 



Eh 
> 
Pi 



CJ CD 

U CD 

ft Eh 

CJ CD 

Eh ft 

CD O 

Eh ft 

CD CJ 

CJ CD 

CJ CD 




Pi CD CJ 

CJ CD 

CJ CD 

CO CJ CD 

Eh < 

, rfj EH 

PC CJ CD 

CD CJ 



H 



Eh ft 

ft Eh 

CJ CD 

CD CJ 

Eh ft 

CJ CD- 

Eh ft 

CD CJ 

Eh ft 

CJ CD 



fci 



CJ CD 

CJ CD 

ft CJ CD 

CD CJ 



CO 



CD CD CJ 
CD CJ 



U 
a) 

I 



Co 



o 



m 



m 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



4/13 



PCT/GB2003/002804 



fa 



CD U 

cd cj 

O CD 

rtj Eh 



H 



fa 



2 




O CD rf! H 

Eh <J a «3 B 

Eh U CD 

CD CJ CJ CD 

CD O CD CD CJ 

CD CJ 



fa 



CJ CD 
Eh 

EH a 

CD O 



Eh CJ CD 

Eh § 

Cti CD S 

CJ CD 

H rf 

PM CJ CD 

CJ CD 

O CJ 

Eh rf 



CJ 
CJ 
CJ CD 
Eh 



CD CD CD 

CD CD 

CJ Eh 

CO CD 




Eh 

CD CJ CD CJ 

CJ Eh rf 

rf CJ CD 




CD CJ 

Pi CD CJ 

CJ CD 

rf Eh 



a 



rf 

CD 



H 



P 



CJ 

8 



eh a g 

Eh CJ 

CD {h rf 
CD Eh 

rf , CD 

Eh £ CD 
CD Eh 
Eh CD 
CJ CD 



Eh Eh rf 
CD Q rf Eh 



CD 
CJ 



gCD U 
Eh 

. CD 

rf Eh 

CD CJ 

CD CD CJ 

cd a 

Eh rf 



i 

M 

B 
o 



CD 



CO 



CD 



CO 



fa 



a rf eh 

CJ CD 

CJ CD 

CD CD CJ 

CD U 

CJ CD 

fa Eh rf 

Eh rf 

CD O 



a 



6 



CM 



rf Eh 

CJ CD 

Eh rf 

rf Eh 

CJ CD 

CJ CD 

CJ CD 

Eh rf 

CD CJ 

CD CJ 



CD CD 

CD 

CJ 

rf CO 



Of 



fa 



CD CJ 
Of rf Eh 
O CD 
H 

EH 

a cd 

E-» rf 

CJ CD U 

Eh rf 

CJ CD 



a 



g 



CD CJ 

Ej rf 

rf Eh 

^ & 

rf Eh 

Eh rf 

CJ CD 

rf Eh 

CD CJ 

Eh rf 

CJ CD 

CJ CD 

Eh rf 

CJ CD 

CD CJ 

rf Eh 

Eh rf 

CJ CD 



fa 



fa 



CJ CD 

CJ CD 

CJ CD 

CD CJ 

rf Eh 

rf Eh 

rf B 

CD CJ 

CD CJ 

CD a 



. U CD 

Pn CD CJ 

U CD 

rf Eh 



o 



H 



fa 




i 



CJ 

^ Eh 

EH 

H H 

CD 
CM CJ 

a 

CD 
* rf 
Eh 
Eh 
H Eh 



rf CO 
Eh 

CD 



CO 



CD 



CO 







$ 


Eh 


fa 








CiJ 






CO 




r i 

vj 








rS 


r * 
kH 






CD 












r ■ 
fcH 








3 




rn 






r._* 








rn 


CD 


v ^ 




















r ■ 










rD 




t 




rn 

ty 


r \ 


> 






rn 


r \ 








rn 


r i 






Mh 










rh 


rn 








til 








cy 


r 1 

y 








r j 
tH 




rn 






r ■ 
fcH 








EH 




rn 










r i 
M 










r Pi 

3 






Hi 














fcH 










i— 1 








rn 

cy 






tH 




r n 

cy 










t^ 


rn 






r \ 
LJ 


rh 




























tH 








rv* 


rn 


r i 

w 


•ft* 




CJ 


rn 






CJ 


rrj 






D 


rf 




Fl, 








r ) 






< 






CD 


CD 


r i 










CJ 
















> 


Eh 


a 








CD 




H 
















B 








CO 












3 






rf 












CD 






CD 


CJ 








Eh 






1 


•CO 


O 










Eh 


3 






O 








v— 1 


Eh 










CJ 


CD 












CM 


a) 




CJ 


CD 






CO 


CJ 


CD 




•s 




Eh 








CJ 


CD 


H 


CO 


CJ 












g 






ril 






m 


CM 


CJ 


CD 








CJ 


S3 


CD 






Eh 














tl 


CO 


CJ 


CD 




«> 




Eh 


a 


CM 












i 


a 




EH 




H 




6 


CD 






U 


CD 




§ 


EH 




CD 




EH 






EH 


Of 






CD 


CJ 




d 




Eh 


< 




■H 


s 














Eh 


a 


}H 




CD 


CJ 




a) 


OJ rf 


Eh 








U 


CD 


>H 


o 




U 


S3 






H 


Eh 






o 






Eh 




e 






CD 






Q 


•=5 


Eh 








CD 


CJ 





in 



o 



f>3 



o 

m 



o 



to 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



5/13 



PCT/GB2003/002804 



u cd 

8 u 

o y 

e-» < 

CD U 

rtj Eh 

CJ CP 

CD CJ 



EH < 

U CD 

CD CJ 

EH j< 



Eh 



U CD 

Eh rtj 

< Eh 

CD CJ 

EH < 

CJ CD 

CJ CD 

Eh fit 

U CD 

CD CJ 
Eh 

Eh fit 

CJ CD 







Eh 




g 


Eh 






CD 


CD 


CJ 


CD 


CD 


CJ 




CD 


CJ 


ft 


CJ 


CD 




Eh 






Eh 


2! 




CD 


£j 


Eh 






CJ 


CD 




fit 


Eh 


> 


CD 


U 




Eh 


< 




CD 


a 


ft 


Eh 






CJ 


CD 




a 


CD 




EH 






Eh 








Eh 


« 




< 




En 




U 


CD 


O CD 


CJ 






Eh 




CJ 


CD 


H 


Eh 


fit 












Eh 


O CD 


U 






Eh 






CD 


a 




Eh 






Eh 




u 


CD 


a 


Eh 






CD 


a 




Eh 






O 


CD 




f$ 




Eh 






3 






CD 






Eh 






< 




CD 


U 




O 


CD 




fit 


Eh 




Eh 


3 


fit 


CJ 


CD 




CD 


CJ 


ft 


Eh 






Eh 






Eh 




O 








Eh 




i 


CJ 






EH 




s 


Eh 




CD 


CJ 


ft 


Eh 






CJ 


CD 




CJ 


CD 


a 




Eh 


3 


Eh 




CJ 


CD 




CD 


CJ 




Eh 


< 




CJ 


S3 




Eh 


fit 


co 










CJ 






Eh 


co 




CD 






CJ 






Eh 


H 


u 








g 












CD 


EH 


CJ 


CD 



Pi 



cd a 

CD CJ 

CJ CD 

rij Eh 




CD 



ft 



OS 



CJ CD Eh 

Eh ml Orf h 

< EH CJ CD 

CD U U CD 

CD CJ CD CD CJ 

Eh fit CD CJ 

CD O CJ CD 

fit Eh j-h" Eh fit 

CJ CD CJ CD 

CD CJ CD U 



Eh CJ CD 

ft CJ CD 
U CD 




rtj Eh 

U CD 

Eh 

U CD 

U CD 

CJ CD 

Eh *C 

CD a 

CD CJ 



ft 



Eh fit 
Eh rij 



Eh rf 

EH ^ 

»< U CD 

CD CJ 

Eh fit 

CO CD CJ 

rtj Eh 

CD CJ 

C EH 

CJ CD 



CD CJ 



CD CJ 
CJ CD* fit Eh 
CJ CD 




H 



O CD CJ CD CJ 
Eh 

CJ CD 

< Eh 
Eh j< 
CJ CD 
•< Eh 
Eh fit 
CD CJ 
CJ CD 

< Eh 
Eh fit 

cj a 




J Eh fit Eh 
U CD ft CJ CD 
CJ CD 



,14 GO CJ 



cd 



6 




CD CJ 

CD CJ 

CJ CD 

f3! EH 



H 



Eh 

CD Q 
CJ CD 

6 



3 
Eh 
CD 



Eh 



CD CJ 
EH fit 

Eh 



CD 



CD CJ « 

< EH 

CJ CD 

fit Eh Eh 

CD CJ 

CD CJ 

CD CJ CD 

Eh < 




CD < Eh 

< a fit Eh 

Eh CJ CD 

CJ CJ CD 

CJ CD CD CJ 

fit CD U 

CJ CJ CD 

Eh Eh 

CD Eh ^ 

CJ CD CJ 




fit 
CD 

| ag 

o CJ 
g H Eh 



H a ^ 

U CJ 

Eh CJ 

CD Eh 

i< CD 

EH & p 

CD Eh 

Eh EH 

CJ EH 




EH fit 

fit EH 

CD CJ « 

& ^ 

CJ CD 

PS Eh Eh 

CD CJ 

CD CJ 

CD CJ CD 

Eh (A 



CD CJ 

CD U 

fit Eh 

rf! EH 

CJ CD 

U CD 

< EH 

cd a 

CD CJ 

CD CJ 




CO CJ CD 
H 

rtj Eh 

CD CD CJ 

CD CJ 

Eh fit 

CO CD CJ 

fit Eh 

CJ CD 

cd a 



fa 



ca CD CJ 

< Eh 

CD CJ 

Of rt{ H 

O CD 

Eh fit 

CO CD CJ 

fit Eh 

■3 EH 

fit CJ CD 

cd a 

CD CJ 

CD CJ 



CD CD U 

CD CJ 

EH fit 

CO CD CJ 



CJ CD 

EH CJ CD 

fit Eh 

CJ CD 

> Eh fit 

CD cj 

Eh rf! 

ft CD CJ 

O CD 

CJ CD 



a cd 

Eh fit 



CO 



3 



CD CJ CD 

CD CJ CD 

fit < CJ CD 

CD CD U 

CD fit Eh 

< W EH 

Eh < Eh 

CD CD O 

CD CD CD CJ 

fit CD a 

6 CD fit E-J 

ft CJ CD 

U CD 



g) ot fit 



O ^ EH 

CJ CD 

CJ CD 

tn EH 

Eh <l 

CD O 

^ CD CJ 

Eh red 

CD CJ 

CD a 



U 

a 

u 

ft 



0) 

ft 



o 



CN 



o 
m 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



6/13 



PCT/GB2003/002804 




EH 


CJ 


cd 












> 


EH 






CD 






E-i 




CM 


CJ 


CD 




U 






EH 






CD 




CO 








TA 












Eh 








> 


Eh 






CD 






CD 






Eh 






S 





CD U 

cd a 

CJ CD 

«3 Eh 



u 



CD CJ 



CO 



HI 



eh <; eh 

CD CO CJ CD 

ft Eh rfl 

O rfj Eh 



CD CJ 
a fcC Eh 
CJ CD 
Eh 

a (3 Eh 



CO 



CO 



u 
a) 



d 



H 



Eh 

Eh ^ 

CD CJ 

ft Eh 

CJ CD 

CJ CD 

Eh ft 

< Eh 

, CJ CD 

CD CD CJ 

Eh Eh < 

CJ CO CJ CD 
Eh Eh 

u . ^ 

CJ >h < Eh 

Eh Eh ft 

i< CJ CD 

CJ H Eh < 

f3 ft Eh 

CD CD CJ 



Eh rfj 

U CD 

CJ CD 

Eh ft 

CJ CD 

CD CJ 

ft eh 

Eh f< 

CJ CD 



Eh 
Eh 
Eh 
Q < . 
CD CJ 
ft Eh 

m ft eh 

CD CJ 



a 



CO CD 



CM 



6 



Oi 



of 



H 



S3 



CJ CD CD CJ 



Pn 



CJ EH 



CM 



CJ CD 

*C Eh 

CD U 

CD CJ 

CJ CD 

Eh ft 

CJ CD 

CJ CD 

Eh ft 

CD CJ 



a ril Eh 

CJ CD 

Eh ft 

CO CD CJ 



a 



Eh ft 
CD CJ 

CJ CD 



CO 



CO 



m ft 
^ cm cj 



Of 



H 



CJ CD U 

O Of < Eh 

CJ CD 

CJ Eh 

Eh Of ft Eh 

Eh CJ CD 

Eh Eh ft 

CD CJ CD CJ 

CJ Eh ft 

ft O CD 

S * £ fcJ 

*E Eh ^ 

CJ CJ CD 

EH >H rtj EH 

CD Eh ft 

CD CD CJ 

ft EH CJ CD 

Eh ft Eh 

CD Eh rf! 

CJ CJ CD 



CD 



ft Eh 

CD CM CJ 

CD CJ 

ft ft 

CD Of ft 

U CJ 



CD CO O 

CD ft 

CD O 

CJ CO CD 

B s 



P4 



CD Eh 
CJ Eh 



CD CJ 
*C Eh 



H 



CO 



O ft Eh 

O O 

Eh ft 

CO CD CJ 

ft Eh 

, ft Eh 

rfj Q CD 

CD CJ 

CD O 

CD CJ 

Pi 

U CD 

CJ CD 

O CD CJ 

Eh ft 

Eh ft 

Eh CJ CD 

f=C Eh 

CJ CD 

H Eh ^ 

ft Eh 



CM 



CO 



CM 



Ei 



CO 



CO 



CM 



CO 



a 



o CD 

CJ CD 

Eh ft 

CJ CD 

CJ CD 

EJ ft 

ft Eh 

O CD 



EH 



H 



CJ 
Eh 



Of 



HI 



CD 



fa 



CD CO 
CD 



Hi 



Of 



u o 

Eh r3 
CD O 



CD 



U 

0) 

a 
» 

CM 



a> 

CJ 

nJ 

CM 



to 



Mn 



o 



to 

CO 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



7/13 



PCT/GB2003/002804 



< Eh 
2 Eh 
U O 
CJ Q 
CD CJ 
CD CJ 
U CD 
Eh rf 
En < 
CD O 

O CD 

Eh rfj 

CJ CD 

f< Eh 

Eh < 

CJ CD 

U CD 

Eh et, 

CD CJ 

U CD 

Eh rf 

CD CJ 

U CD 

CD CJ 

CD CJ 

b§ 

CD O 

< Eh 

a CD 

Eh 

U CD 
Eh f=C 

cd a 

Eh < 
U CD 

< EH 
Eh ft. 
CJ CD 

Eh rf: 

CD CJ 

CJ CD 

< Eh 
Eh < 
CJ CD 

CD CJ 
Eh 
Eh 
Eh 

EJ ■ 

< Eh 

CD U 
Eh 
f3 Eh 

cd a 

EH < 

O CD 
U CD 
Eh 

CJ CD 

CD CJ 

Eh < 

CJ CD 

Eh < 



Eh 




Eh i3 

CD CJ 

< Eh 

a cd 

ft Eh 

CD CJ 

CD CJ 

CD CJ 
Eh 



CD U 

W CD U 

a eh 

Eh CJ CD 

CJ CD 

ft Eh 

CD CD CJ 

CD U 

CD CJ 



3 B CO Eh .< Ot 
Eh 

Eh . 

CDCJCDCDCJCDCDCJ 



ft Eh 

f3 EH 

CJ CD 

CJ CD 



CJ CD 
CD O 



U CD CD CJ 



EH J* 



CJ Pn CJ CD 



EH 



ft Eh 

O CD 

ft Eh 

CJ CD 



Pi 



< Eh 

Eh ft 

CO CJ O 

Eh rf 

Eh ft 

Eh ft CJ CD CO CD U 

Ej f< 

W ^ Eh 

CJ CD 



U CD 

> CD CJ 
CD CJ 



CD 



CD CJ 
CD CD CD CJ O* 

CJ 



3 



co 



Eh ft 
CD CJ 



CD CJ 

6 Eh 
CD 



cd a cj cd a cd cj 



CJ CD 
CJ CD 
Eh 
CD 



O CD 



CD 



fa 



fa 



Eh ft Eh rfj 

U CD ft CJ CD 

CJ CD U CD 

Eh f3 Eh 

CJ CD CX Eh 



EH 



Eh 



CD 



6 

CJ 
Eh 

6 



s 



s 



CD 



Eh rfj 
EH ^ 



CD U 

Ph CD CJ 

CJ CD 

ft Eh 



H Eh 



CJ CD 
< Ej 



fa 



CD 



CJ CD ft Eh 

Eh r3 Oi rfj Eh 

ril Eh CJ CD 

CD CJ CJ CD 

CD CJ CD CD CJ 

Eh CD CJ 

CD U CJ CD 

ft Eh fa Eh ft 

CJ CD H r< 

CD CJ CD CJ 



CD 
fa < 



6 



fa 



ft 



CJ 



CO 



CD CJ 

Eh < 

CD CJ 

fit Eh 

CJ CD 

Eh rf 

Eh *C 

Eh fit 

CD CJ 

CJ CD 

fit Eh 

O CD 

Eh < 

rtl Eh 

U CD 

CJ CD 

CJ CD 

Eh ft 

CD O 

CD CJ 

CD CJ 

CD a 

Eh fit 

CD O 



6 



Eh O CD 

< Eh 

. Eh fit 

Pn CD CJ 

CJ CD 

EH < 

ft O CD 

O CD 

Eh ft 

„ Eh 3 

fa 

EH 

Eh *q 

^ S 

Eh fit 

CD U 

CO CJ CD 

Eh fit 

Eh f< 

P rfj Eh 

CD CJ 

CD CJ 

O <C Eh 

CJ CD 

rtl Eh 

a ft Eh 

O CD 

Eh (3 

CJ O CJ 

Eh (3 

CJ CD 



8 



fa 



EH fit 

cj a 

CJ CD 

Eh < 



3*3 



H 



Eh 



&h rf 

fit Eh 

0 CD 

CJ CD 



CD CJ 

Pi CD CJ 

O CD 

rfj Eh 



H 



P 



3 



eh a ^ 

u o 

Eh CJ 

CD >h rtj 

CD Eh 

< „ tD 

Eh & CD 

CD EH 

Eh Eh 

CJ CD 



Eh *S 

Eh CJ CD 

fit Eh 

CJ CD 

Eh < 

Eh ^ 



J Eh 
Eh 



Eh 



CO 



CD 



6 



Eh 

CD CO 
CJ 

Eh ft 



Ph 



CD CD CD 



H 



H 



6 



1 



CJ 



§ 



rfj «< CJ 
CD CD 



CD CD 
CD CD CD 
< CD 



co a < 

& CJ 
O CJ 

a h ej 

o < 

% 

CD 



Q 



Eh 
O 
Eh 
CD 

CD 
Eh 
O 



CJ CD 

EJ < 

< Eh 

CD U 

CD U CD 




< Eh 

O CD 

eh m; o 

^ Eh 

O CD 

CJ CD CO 

CJ CD 

Eh <: 

CD CJ Q 

CD CJ 



O 

O Of 
U 

Eh Ot 



fa 



H 



SEh 
CD P 

U CD 

< Eh 

Eh Eh 

CD CJ 

CD O 

Eh ft, CD 

CD O 

< Eh 



§ 

4J 

u 
ft 



u 

CO 

a 

M 
n) 
ft 



wo 



O 



O 

CO 



wo 

CO 



wo 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



8/13 



PCT/GB2003/002804 



3 



ft H 

ft H 

CJ CD 

CJ CD 

CD a 

CD CJ 

CJ CD 

Eh ft 

Eh f2 

CD CJ 

cj cd 
(< 

Eh 

Eh < 

O CD 

U CD 

eh pj; 

CD CJ 

cd a 

CD CJ 

a u 

CD CJ 

EH < 

CD O 

< Eh 

CD CJ 

cd a 

ft Eh 

CJ CD 
Eh 

ft H 

CJ CD 

Eh ft 

CD O 

Eh rij 

CJ a 

< Eh 

Eh ft 

CJ CD 

Eh ft 

CD O 

CJ CD 

< Eh 
Eh rf! 

a a 

CD cj 

Eh 
Eh 

f *i 

Eh 

rfj Eh 

CD CJ 

rf} Eh 

3 Eh 

CD CJ 



Eh < 

U CD 

a cd 

ft Eh 
Eh 

U CD 

CD CJ 

Eh ft 

O CD 

Eh < 





CD 

ft a 

Eh 
U 



Eh 
< EH 
CJ CD 
CJ CD 



Ph CD CJ 

ft Eh 

CD CJ 

CD CD CJ 

CD CJ 

Eh ft 

Eh ft 

ft Eh 

O CD 

CO CD CJ 

ft Eh 

CD CJ 

a ft EH 

CJ CD 

Eh rfl 

to CD CJ 

' Eh 
Eh 

ft CJ CD 

CD U 



CJ CD CD U 

CD CJ 

CJ CD 
Eh 



ft 
CJ 

Eh fa 



a) 



CJ CD 

U CD 

ft Eh 

O CD 

EH ft 

CD CJ 

P* CD CJ 
U CD 
a CD 

Q ^ EH 

CD CJ 

ft Eh 

CD CD CJ 

CD CJ 

< Eh 

S6 

Eh «< 

CJ CD 
CO 

ft S CD 

CD CJ 

Eh ft 

CO CJ CD 

Eh ft 

CD CJ 

Eh ft 

CJ CD 



O CD 

EJ < 

i«C Eh 

cd a 



rfj Eh ft 
CD Ai CJ CD 



U CD 

CO U CD 

Eh ft 

CJ CD 

CO CJ CD 

EJ ft 

ft EH 

Pj CJ CD 

CJ CD 

Eh ft 

CO CJ CD 

Eh ft 

CD CJ 

Ol ft Eh 

U CD 

CJ CD 

EH CJ CD 

ft Eh 

CD CJ 

Eh ft 

ft EH 

CD CJ 

O rtj Eh 

CJ CD 

CJ CD 

Eh <C 



H 



a 



EH 

U CD 
ft Eh 
CD CJ 




CD CJ 
Pi CD CJ 
CJ CD 
Eh 



CD O 
ft Eh 





<y ft 

CJ 

EH 

to CD 



Eh Eh 
CD to CJ CD 




U 

0) 

Pi 



O 
rH 

i 

a) 

g 

CD 



a) 
to 



fa 



fa 



&1 



Pi 



fa 



IP 
I 



o 

CN 



o 

m 



m 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



9/13 



PCT/GB2003/002804 



cd cj 

u cd 

u cd 

CD CJ 

CD CJ 

CD CJ 
EH 

d En 

CD CJ 

Eh rij 



CD U 

U CD 

CJ CD 

Eh ft 

U CD 

f3 Eh 

CD U 

< Eh 

CJ CD 

CD CJ 




< 

CJ CD 

CD U 

U Eh rtj 

CD U 

Eh rtj 

>h O CD 

< Eh 
Eh isfi 
Eh ^ 

>h f< Eh 

Eh ft 

> gS S3 

Eh rfj 

CD O 

CD CJ 

CJ CD 

CD CJ 

Eh U CD 

CJ CD 

< Eh 

POO 
Eh 

CD CJ 

W CD CJ 

ft Eh 

CD CJ 

rtj CJ CD 

CJ CD 

Eh paj 

CD CJ 

CJ O 

CD CJ 

Eh rij 

CJ CD 

CO CJ CD 

CD CJ 

g g 

S ft Eh 



CD U 

Eh ft 

CD CJ 

f< Eh 

CD CJ 

& ^ 
Eh ft 

CJ CD 

Eh 

CD 

CD CJ 

CD CJ 

ft Eh 

ft Eh 

CD CJ 

CD CJ 

CD CJ 

5S 

CJ CD 







Eh 










CD 


a 
























Eh 








Of 


i 


Eh 












Eh 




CD 


CJ 




CJ 


CD 


CO 


U 






CD 


CJ 




EH 






Eh 






CJ 






O 


0 




Eh 










> 










CD 


u 


g 




CJ 


CD 








Eh 


CJ 


CD 




CD 






ft 


Eh 




Eh 




CJ 


S3 




CJ 


0 


> 


Eh 


6 


Eh 


CD 


o 




CD 








CD 




CD 


CJ 










Eh 








CD 




CJ 


CD 


















EH 




CJ 


CD 




1 


Eh 


H 


CJ 


CD 






CJ 




% 


Eh 






Eh 






Eh 




5 


Eh 


CD 


CD 


U 


CO 


CJ 


CD 




CD 


CJ 




CJ 






CD 


CJ 




EH 




a 


*C 


Eh 






s 




CJ 


CD 


S3 




Eh 




CJ 


CD 






Eh 


CD 


CD 


CJ 


P 


1 


CD 




a 


CJ 






Eh 




CD 


CJ 






CJ 


IS 


CD 


CJ 




CJ 


CD 




Eh 


0 


CD 


CJ 










CJ 


CD 


>H 


% 




CO 


CJ 


CD 




Eh 


% 




CJ 








B 




Eh 


*u 




3 




H 


CJ 


S3 


p 


CD 


CJ 










Eh 










fa 


Eh 




Eh 




CD 




Eh 






CJ 


CD 




Eh 








Eh 


p 




Eh 


fa 


0 


CD 




CD 


CJ 


EH 






CD 


CJ 




Eh 






< 


Eh 




CD 






CD 


CJ 




EH 




fa 



CO CJ CD 

Eh ft 

CJ CD 

P rtj H 

CD CJ 
Eh 

CJ CD 

CD CJ 



CJ 



>H 



CJ CD 

CD CJ 

Eh ft 

CD CJ 

Eh *3 

O CD 

< Eh 
Eh 

Eh g 

ft EH 



fa 



CD O 

ft Eh 

f3j CJ CD 

CD O 

Eh ft 

CJ CD CJ 
Eh 

U CD 

CO CJ CD 

Eh ft 

CJ CD 

>i eh <: 

CJ CD 

Pi CD U 

CJ CD 

CD CJ 

A Eh <: 

CJ CD 

CJ CD 

CO CJ CD 

Eh ft 

CD CJ 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh ft 

CJ CD 

ft 

CJ CD 

CD CJ 

O* ft Eh 

CJ CD 

^1 E~j 

> Eh ril 

CD CJ 

CD CJ 

A Eh ft 
Eh 




O 

, Eh 
< CJ 
CD 

P S 
CD 
Eh 



CJ P CD CJ 

CD CJ 

ft Eh 

fa CD CJ 

CJ CD 

cj cd 

CD , CD CJ 

ft CJ CD 



CD 
U 



CO 



CD CJ 

CJ CD 

CD CJ 

ft Eh 

rtj Eh 





CD 


CJ 


s 




B 


EH 


CO 


CJ 






CD 








B 


IS 
















s 




f. 


> 


CO 


TC 


AG 






Eh 
Eh 






CJ 


9 


a 




CD 




> 




ft 




CD 








CD 


CJ 




EH 








CD 


CJ 


i-h 1 


CJ 


CD 





CD CJ 

CO CJ CD 

Eh ft 

U CD 

>Bc1 

CJ CD 



CJ CD 

ft Eh 

O CD 

Eh ft 

CD CJ 

CD CJ 

Eh <1 

CJ CD 

a cd 

cj cd 



u 

■H 



IS CD 
Eh 
CD 

w ft 

i-3 Eh 
CJ 
Eh 
CD 



CJ CD 

CD CD CJ 

CD CJ 

ft Eh 

SCJ 
CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh ft 



CD 



CD 
CD 



CD 



CD 
CD CD 

S3 

es 

cj 



a 

6 >h 

Eh 
CJ 

Eh ^ 

^ 
CD 

0 

O 

CJ s 
Eh 

CJ ^ 
CJ 

a 



EH 

u 

CD 
Eh 
CJ 
CD 
U 

6 



Eh 

. EH 

CD CJ 

CD CJ 

CD CJ 

ft EH 

CJ CD 

CJ CD 

CD CJ 

CD CJ 



CD 



a 

CJ 



ft 



Eh CO EH 



CD 
CD 



, Eh ft 

ft CJ CD 

CD CJ 

CD CJ 

ft Eh 

CJ CD 

CJ CD 

Ph CD CJ 

CJ CD 

CJ CD 



a 



S 0S 

P CD CJ 

CJ CD 

, CD CJ 

Ph CJ CD 

CJ CD 

CJ CD 

CO Eh < 



a 




u 
a) 

•H 



CJ Eh ft 

CD CO CD CJ 

CD rfi Eh 

CJ Eh ft 

CJ CD CD CJ 

U CD CJ 

Eh ft Eh 

Eh « Eh 



CD CJ 
Eh ft 
CD CJ 



n y w 

Eh 

EJ g 

< Eh 

CD CD U 

CD U 

U CD 

CO CJ CD 

Eh ft 

CJ CD 

. CJ CD 



Ei W < B 



CJ o 

CD CJ 

^ Eh ft 

CJ CD 

CJ CD 

CO CJ CD 

Eh ft 

CD CJ 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 
£h 

a cd 



ft* 



3 



CJ Eh 
< >h rij 



O CD 

CD CD CJ 

cd a 

ft Eh 

CD CD CJ 

CD CJ 

CD O 

CD CD CJ 

CD CJ 

Eh ft 

CO CJ CD 

Eh ft 

CD CJ 

W *3j Eh 

CD CJ 

CD CJ 

•-h" Eh ft 

Eh < 

CD CJ 

Eh rtj 

CJ CD 

CD CJ 

Ot ft Eh 

CJ CD 

CD CJ 

> Eh < 

CD U 

CD CJ 

W ft Eh 

CD CJ 



CD 



Eh ft 

> 

CD CJ 

CJ CD 

Eh CJ CD 

ft Eh 

CJ CD 

> Eh ft 

CD a 

CD CJ 

iA &H ft 

CJ CD 



0 

CJ 
CJ 

% 



H 



< EH 

CJ CD 

ft Eh 

CD CJ 

CJ CD 

CD CJ 

CJ CD 

CJ CD 

CJ CD 

Eh ft 

U CD 

Eh ft 

ft Eh 

U CD 

CJ CD 

ft Eh 

CJ CD 

Eh ft 

Eh ft 

CD CJ 



CD 



fa 



CO 



P 



3 



cd a 

U CD 

U CD 

Eh ft 

U CD 

rf! Eh 

CD CJ 

ft Eh 

CJ CD 

CD a 

CJ CD 

< Eh 

Eh ft 

CD CJ 

rf! Eh 



CJ CD 

ft Eh 

CD CJ 

CD CJ 

CD O 

Eh ft 

CD a 

CD CJ 

Eh ft 

CJ CD 



8 



Eh < 
CD U 
CD CJ 



i 

CD 
CD 
CD 



in 

I CD 

& w 

CN 

i 

•H h3 

o 

CD 

fi a 

o CD 

a > Eh 

O CD 

H CD 

A CD 




CD ft 

ft ft CJ CD 

CJ CD CJ 

Eh CD U 

a a ft Eh 

CJ CJ CD 

CJ CD 

Ptf CD CJ 

CJ CD 

ft CJ CD 



CD 

a 

Eh 
CD 

a 

6 

CJ 
Eh 
CJ 



> Eh 

CD CJ 

CD CJ 

Eh c2 

CD CD CJ 

CD CJ 

CD CJ 

Eh ft 



CN 



o 

CO 



IT) 
CO 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



10/13 



PCT/GB2003/002804 




<H U CD 

cd cj 

B fit 

CJ CD CJ 

Eh rfj 

o cd 

J* < H 
Eh 

cd cj 

< cj cd 

CD CJ 

cj cd 

Eh a CD 

(< Eh 

CJ CD 

P Eh 

CD CJ 

CD U 

H fit Eh 

CD CJ 

CJ CD 

rfj CJ CD 

CD CJ 

Eh <C 

Pi CD CJ 

CJ CD 

CD CJ 

hI H fl! 

U CD 

CJ CD 

CO CD CJ 



CD CJ 

CO CJ CD 

Eh < 

CJ CD 

U CD 



Eh 



CJ CD 

fit Eh 

CJ CD 

Eh fit 

CD CJ 

cd a 

Eh fit 

a cd 

CJ CD 

CJ CD 



Eh 

. En 

CD CD CJ 

CD CJ 

CD U 

a < eh 

CJ CD 

CJ CD 

CD CD CJ 

CD CJ 



, CJ CD 

Pi CD CJ 

CJ CD 

CJ CD 

W CJ CD 

EH < 

CJ CD 

H Ej g 

fit Eh 

CJ CD 

EH CJ CD 

fit Eh 

CJ CD 
fa Eh 

gg 



U 



Eh Pi CD 



CD 



fa 



CD CO U 
Eh 



CD CJ 
Eh 

O CD 



CD CD 



3 



5^ 



*3 



CD CJ CD 
fit Q tfJ Eh 



CD W rij 



Pj 



CD CD CD 



CD CD 

< o <: 

U CJ 

u 

U CO CJ 

CJ Eh 

CJ CJ 

U > Eh 

fit CD 

CD CD 



Eh Pi CD 



CJ CO CD 



d 



g 



Eh 



CD 



EH 



fa 



CD 
W «3 



Eh 



cd > eh 

CJ CD 

Eh CD 

CD ^ CD 

CJ Eh 

U CD 8 

CJ CD 

Eh CD 

CJ Eh 



fa 



CD 



Pi 



<C Eh 
Eh ^ 
co CD CJ 

Eh . fit 



CD 



g 



fa Eh 

&H g 

fit Eh 

O CD CJ 

CD CJ 

CJ CD 

CO CJ CD 

Eh fit 

CJ CD 

CJ CD 

CD O 

fit Eh 

< U CD 

CD CJ 

Eh fit 

CJ CD CJ 

Eh fit 

CJ CD 

CO CJ CD 

Eh fit 

a cd 

h! EH fit 

£tj CD CJ 

CJ CD 

CD CJ 

Hi EH fit 

a cd 

CJ CD 

CO CJ CD 

Eh fit 

CD CJ 

CD CD U 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh fit 

CJ CD 

P< 



CO 



Pi 



CO 



CD CJ 
fit Eh 



CO 



S2 
CJ 



CD CJ 
Eh «3 
CJ CD 



6 



CJ CD 

CD CD CJ 

cd a 

fit Eh 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh rf{ 



5*4 



6 



S 



CJ CD 

CD CJ 

a fit eh 

CJ CD 

CD CJ 

cd a 

H rf! Eh 

CD U 



CO 



fa 



CJ CD 

EH < 

CO CJ CD 

Eh fit 

Eh rf! 

CO CJ CD 

EH fit 

EH 3 

O f=S E-J 

CD CJ 



CD 



fa 



CO 



Q 



fa 



CD U 

CJ CD 

CJ CD 

Eh fit 

CJ CD 

fit Eh 

CD CJ 

fit Eh 

CJ CD 

CD CJ 

CJ CD 

< Eh 

Eh ft 

CJ CD 

fit Eh 



CD 



EH 



EH fit 

fit CJ CD 

CD CJ 

fit Eh 

CO CJ CD 

Eh fit 

CJ CD 

> Eh rfj 

CD CJ 

CD CJ 

CD CJ 

H *C Eh 

CD U 

Ufl Eh 
Hi Eh 

U CD 

CD 

CD CJ 

CD CJ 

fit Eh 

fit Eh 

CD CJ 

CD CD CJ 

CD CJ 

6EH 
CD 

CJ CD 

A Eh ft 

fit CJ CD 

CD CJ 

CD CJ 

a < eh 

CJ CD 

CJ CD 

Pi CD CJ 

CJ CD 

CJ CD 



fa 




u 



td 

Pi 



o 

rv3 



o 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



11/13 



PCT/GB2003/002804 



cj cd 

Q < Eh 

CD O 

cd a 

W rtl Eh 

CD CJ 

CJ CD 

f< U CD 

CD CJ 

Eh f< 








Eh 




a 


CD 






Eh 






S3 


CJ 


Eh 




CD 


TT 






TT 






CD 


CD 


CD 


U 


CO 


u 






CD 


CJ 




EH 




S3 


U 








a 


f3 


Eh 




< 


Eh 
£h 




CJ 


CD 


125 




CJ 


CD 




a 


CD 


CD 


CD 


CJ 


Q 




EH 




CD 


CJ 


CD 


a 




CD 


U 


P* 


CJ 


CD 




CD 


CJ 


CD 


CJ 




Eh 




CJ 


CD 




a 


g 




CJ 




< 


I 


CO 


CJ 


1 




Eh 






EH 


< 




a 


CD 








< 


EH 




CJ 


S3 


P 




H 


CAT 


IS 

CD 


fa 


TTG 


U 


EH 


CJ 


CD 




EH 






s 


Eh 


J3 










S3 

s 






fa 


Eh 

EH 




a 

S3 


CT 




CD 


CJ 


W 


g 


Eh 










U 




CJ CO CD 
EH < 
CD CD 
CJ CD CD 
CD 
Eh 

rij CD CD 



rij U CD 

CD CJ 

CJ CD 

Eh CJ CD 

< Eh 



CJ CJ CD 

Eh D f$ Eh 

CJ CD O 

CJ CD U 

CJ fa f3 Eh 

f< CD O 

CJ U CD 

cj rfj a cd 

Eh Eh f< 




a) 

n 

(d 

Pi 



CJ Eh rfj 

CD >h rtj Eh 
CD Eh 
CJ H 

O M Eh 
CJ 




Pi 



fa Eh tfj 
Eh 

< Eh 

CD CD CJ 

CD CJ 

U CD 

to U CD 

Eh < 

CJ CD 

CJ CD 

CD U 

, < Eh 

rf! CJ CD 

CD CJ 

Eh rfj 

CJ CD O 

Eh 

CJ CD 

CO CJ CD 

Eh < 

CJ CD 

hi Eh (3j 

CJ CD 

Pn CD CJ 

CJ CD 

CD CJ 

^ Eh < 

CJ CD 

U CD 

CO CJ CD 

H < 

CD O 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 
Eh 



Eh fa rfj 



a cd 



CJ CD 

CD CJ 

a & Eh 

CJ CD 

*C Eh 

CD CJ 
hi B 

^ Eh icq 



Q) 

•H 
H 

&» 
m 



oo 

IT) 

i 

CNJ 



CJ CD 

CD CD CJ 

CD CJ 
Eh 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

cd a 

Eh rf) 

CO CJ CD 

Eh < 

CD CJ 

fa < Eh 

CD CJ 

CD CJ 

J Eh 

EH < 

CD CJ 

Eh < 



CD 



OJ 



CJ CD 

CD CJ 

6Eh 
CD 

CD CJ 
Eh 

CD a 

CD CJ 

H K< EH 

CD CJ 



EH 



S3 H y 



Eh fa 

CD 

Eh 

CJ *3 



Eh 

EH f3 

CD CJ 

CJ CD 

CD CJ 

Eh i«C 

CJ CD 

CD CJ 

Eh < 

CJ CD 



CD 



CD 



fa Eh rfj 

Eh *3 

rtl Eh 

CD CD CJ 

CD CJ 

CJ CD 

to CJ CD 

Eh < 

CJ CD 

, CJ CD 

CD CJ 

<SS 

CD CJ 

Eh < 

CJ CD CJ 

Eh rtJ 

CJ CD 

CO CJ CD 

Eh ft, 

CJ CD 
J EH 

U CD 

CD CJ 

U CD 

CD CJ 

A Eh r3 

U CD 

U CD 



CD CJ 

CJ CD 

CJ CD 

CD CJ 

CD CJ 

CD CJ 

rfj Eh 

rf! Eh 

CD CJ 

Eh rfj 

CD CJ 

CJ CD 

O CD 

Eh rfj 

CJ CD 

< Eh 
CD CJ 

< EH 
CJ CD 
CD CJ 



CJ CD 

>i Eh 
Eh 

. CJ CD 

Eh 

< 

Eh U CD 

< EH 

CJ CD 



CO 



Q 



6 



CO 
CD 
CD 

Pi 



u 

pi 

iH 



CD 



CD 



ro CD 



TJ co 



fa 



ID 

1 



•H hi 

u 

6 



fa 



CJ CD 

Eh < 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

Eh rtj 

CJ CD 

CJ CD 

a cj 

*C Eh 

CJ CD 

< Eh 
Eh -f!d 
CD CJ 
CD CJ 

CJ CD 

CD CJ 

cd a 

ri{ Eh 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

Eh < 

cj cd 

Eh < 

CD CJ 

< Eh 
CD CJ 
CD CJ 

E-i 

CD U 

Eh <3 

O CD 

CD CJ 

rij Eh 

CJ CD 

CD CJ 

Eh rtl 

CD O 

CD CJ 

rtj Eh 

CD CJ 



CO CD CJ 

< Eh 
Eh rij 

CD CD O 

CD CJ 

Eh <: 

CD CD CJ 

CD CJ 
Eh 
CD 

CD CD 

CD CJ 

Eh t$ 

CO CD O 

rij Eh 

Eh 

H EH rtj 

ril EH 

eh <: 

rfj CJ CD 

CD CJ 

< Eh 
CO CJ CD 

Eh < 

U CD 

cd a 

|S CD CJ 

<C Eh 

^ Eh < 

U CD 

EH *C 

a CJ 

CD 

CD CJ 

CD CJ 

^ Eh 

3 Eh 

CD CJ 

CD CD CJ 

CD CJ 

rtj Eh 

CM CJ CD 

CJ CD 

^ ^ < 

rf| CJ CD 

CD CJ 

CD CJ 

a t4 Eh 

CJ CD 

CJ CD 

Pi CD CJ 

CJ CD 

CJ CD 

CD CJ 

& CD O 

Eh fiC 
Eh 

CO U CD 

CD CJ 

Eh < 



in 



CD 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



12/13 



PCT/GB2003/002804 



6 



cd 



En 



Q 



EH rf! 
Eh 3 



3 



H 



fa 



cd 



CO 



CD CJ W 

fit Eh 

CJ CD 

CD CJ Eh 

Eh <C 

CD O 

Eh rfj CD 

CJ CD 

CJ CD 

eh 

CJ CD 

Eh < 

CJ CD Pi 

Eh ^ 

CD CJ 

CJ CD CD 

CD CJ 

Eh fit 

CJ CD H 

CJ CD 



Ot 



CD U 

CJ CD 

CJ CD 

CD CJ 

CD CJ H 

CD CJ 

S Eh US 

CD CJ 

Eh < 

CD CJ rfj 

U CD 

CJ CD 

Eh ^ U 

CJ CD 

fit Eh 

CD CJ >h 

< Eh 
CJ CD 
CD CJ , 

>h 

CJ CD 

< Eh . 



CD CJ CJ 
CD CJ paj CJ 



s 



CJ 
CD 
CJ 

fit CD U 
CD CO U CD 

CD Eh rf! 

cj cj q 



a cd 

fit Eh 

CJ CD 

Eh fit 

CD a 

CD CJ 

Eh fit 

CJ CD 

cj a 

CJ CD 

fit Eh 

3 Eh 

CD CJ 

CD CJ 

CD CJ 

fit Eh 

U CD 

CJ CD 

CD U 

CD a 



CD 



CJ 



CJ CO CJ 



5 



v X 

9 



CD Pi CD 



S 



CD 



CD CJ 
*C Eh 



CD 



fa EH 

Eh ^ 
Eh 

CD CD CJ 

CD CJ 

CJ CD 

CO CJ CD 

Eh *H 

CJ CD 

CJ CD 



3 



CJ rt) CJ CD 
CJ CD O 
CJ Eh rfj 



, u CD 
h3 Eh < 
CJ CD 



Pi CD CJ CD CJ 



CD 



CO CJ CD 

Eh rf 

CD O 

CD CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh fit 

CJ CD 

ft 



CO 



s 

CJ 
CJ 

6 



CJ CD 

fit Eh 

CJ CD 

Eh fit 

CD CJ 

CD CJ 

Eh fit 

CJ CD 

a cd 

CJ CD 



u 

Q) 

•rl 
H 



Eh rf 
CD CJ 
CD CJ 



t-i 

H 

in 
i 

►a 

% 



a cd 

CD CD CJ 

CD U 

< Eh 

O CD CJ 

CD CJ 

CD CJ 

CD CD CJ 

CD CJ 

Eh ft 

CO CJ CD 

Eh fit 

CD O 

fa fit Eh 

CD CJ 

CD CJ 

hh" Eh 
Eh 
CD 
Eh 



CD 
CD CD 



2 



Eh 
Eh 

CD CJ 

CD CJ 

CD CJ 

fit Eh 

CJ CD 

CJ CD 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

EH fit 

CJ CD 



CJ CD 

fit Eh 

CD CJ 

Eh < 



6 



fa 



ft 



8 



CO 



Pi 



CJ 



CD 
CD CJ 



W f£ Eh 
CD CJ 



CJ CD 

H < 

< Eh 

U CD 

CJ CD 

fit Eh 

CJ CD 

Eh fit 

Eh < 

CD CJ 



Pi 



CD 



ft 



CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CJ CD 

Eh < 

CD CJ 

CD CJ 

CD CJ 

CJ CD 

CJ CD 




fa Eh fit 

fit Eh 

CD CD CJ 

CD CJ 

CJ CD 

CO CJ CD 

EH fit 

cj cd 

CD CJ 

rfj 3 CD 

CD CJ 

Eh < 

CJ CD CJ 

Eh fit 

CJ CD 

CO CJ CD 

Eh fit 

CJ CD 

\1 Eh fit 

CJ CD 

Ph CD CJ 

CJ CD 

CD CJ 

a Eh fit 

CJ CD 

CJ CD 



CJ CD 

^ ^ Eh 

Eh *3 

CD CJ 

Eh CJ CD 



CO 
CD 
CD 

ft 

a 



u 
a> 

CD 

♦H 

m 

H 

T3 CO 

m 

43 fa 

CN 



EH 

■H hh 1 



U CD 

EH f< 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

CD CJ 

Eh fit 

CJ CD 

CJ CD 

CD CJ 

< Eh 
CJ CD 

& % 
Eh 

CD CJ 

CD U 

Eh fit 

Eh fit 

CJ CD 

CD CJ 

cd a 

fit EH 

CD CJ 

CD CJ 

cd a 

CD CJ 

CD CJ 

Eh ft 

CJ CD 

Eh fit 

CD CJ 

^ Eh 

CD CJ 

CD CJ 

Eh fit 

EH r3 

CD CJ 

Eh fit 

CJ CD 

CD CJ 

< Eh 
CJ CD 
CD CJ 
Eh fit 
CD U 
CD CJ 

ci B 



Eh rt^ 

CD CD O 

CD CJ 

*C Eh 

co a cd 

Eh fit 

CJ CD 

CD CJ 

sou 

Eh *C 

CD CJ 

fa *S Eh 

CD CJ 

fit Eh 

J Eh fit 

CJ CD 

g 



CD 



CD 



CD CJ 

CD CJ 

ret! Eh 
Eh 

CD CJ 

CD CD CJ 

cd a 

fit Eh 

ft u o 

cj a 

EH fit 

fit CJ CD 

CD CJ 

CD CJ 

O fit Eh 

CJ CD 

CJ CD 

Pi CD CJ 

CJ CD 

CJ CD 

> Eh fit 

CD CJ 

CD CJ 

£ CD CJ 

Eh fit 

CD CJ 

PJ CD CJ 

fit Eh 

CD CJ 

Eh ft 



U 

s 

-p 



o 

CN 



O 



o 



SUBSTITUTE SHEET (RULE 26) 



WO 2004/003019 



13/13 



PCT/GB2003/002804 



Eh 



CJ CD 

Q < En 

CD CJ 

cd cj 

W < ^ 

CD CJ 

U CD 

ft CJ CD 

CD U 

H < 



CD U 

U CD 

Eh f$ 

U CD 

Eh ft 

CD U 

CJ CD 

CJ CD 

ft Eh 

U CD 

Eh < 

CD U 

CD CJ 

Eh ft 

CJ CD 

U CD 

CJ CD 



Q 



to 



H 



, CD U 

paj cj o 

CD U 

CD CJ 

CD CD CJ 

CD CJ 

CD U 

CD CD U 

CD CJ 





rf 








EH 




CD 


CJ 




CD 


CD 


CJ 




CD 


CJ 


CD 






< 
















ft 




CD 


u 






CD 


CJ 




CJ 


CD 










> 






CJ 




* 




Eh 






Eh 




co 




Eh 






to 














Q 




CJ 


CD 




Eh 




CD 








g 













CD CJ E-J rtj 

cj CD a *C Eh 

CJ CD CJ CD 

CD CJ CD U 

CD CJ Pi CD CJ 

CD CJ 

3 £ m 

CD U 

Eh < 




Ptj Eh 

ft 
CD CD 
CD 
CJ 
CO CJ 
EH 
CJ 

ft u 

CD 
Eh 

O CD 
Eh 
CJ 

W CJ 



3 !H 

Eh 

CJ 

CJ >H 

CD 

CD 

rij Eh 

CD 

CD 

CJ CO 

Eh 

CD 

CJ CD 



CD CJ 

CJ CD 

CJ CD 

Eh < 

CJ CD 

ft Eh 

CD U 

ft EH 

CJ CD 

CD CJ 

O CD 

Eh ft 

CJ CD 

Eh 

ft Eh 

CJ CD 

ft Eh 

CJ CD 



ft CJ CD 

CD CJ 

Eh ^ 

CJ CD 0 

Eh < 

CJ CD 

>h < Eh 
Eh 
Eh 

■ < 

<J Eh 

CD CJ 

ft CJ CD 

CD U 

CJ CD 

Eh CJ CD 

ft Eh 



6 



CD CJ 

f< Eh 

Eh ft 

cd a 



rtj CD 
CD 



CJ CD 

Q ft Eh 

CD CJ 

CD CJ 

CD CJ W ft Eh 

ft CD CJ 

CD CJ CJ CD 

CD CJ ft CJ CD 



CD CJ 

03 U CD 

Eh rfj 

CJ CD 

Eh rti 

CD CJ 

CJ CD 

CJ CD 

ft Eh 

CJ CD 

Eh *C 

CD CJ 

CD CJ 

Eh ft 

CJ CD 

CJ CD 

CJ CD 




to EH 
EH 
CD 

a rfj Eh 

CJ CD 

CD CJ 

Ph CD CJ 

< Eh 

Eh rfj 

CJ CD 



CJ 
CD 
O* ft 
CJ 

> ^ 
CD 

CD 

to 

CD 



CD 
CJ 
Eh 
CD 
CJ 

6 

CJ 
EH 
CJ 



ft CJ CD 
CD CJ W Eh ft 
CD CJ 





SUBSTITUTE SHEET (RULE 26) 



INTERNATIONAL SEARCH REPORT 



lnt< >na! Application No 

PC77GB 03/02804 



A. CLASSIFICATION OF SUBJECT MATTER 

IPC 7 C07K16/24 C07K16/18 
C12N15/63 C12N15/62 



C07K 16/28 
C12N15/13 



A61K39/395 C07K16/46 



According to International Patent Classification (IPC) orto both national classification and IPC 



B. FIELDS SEARCHED 



Minimum documentation searched (classification system followed by classification symbols) 

IPC 7 C07K 



Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched 
Electronic data base consulted during the international search (name of data base and, where practical, search terms used) 

EPO-Internal , BIOSIS, WPI Data, PAJ, MEDLINE, EMBASE 



C. DOCUMENTS CONSIDERED TO BE RELEVANT 



Category 



Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



A 



X 
Y 



ELS CONRATH K ET AL: "Camel single-domain 
antibodies as modular building units in 
bispecific and bivalent antibody 
constructs " 

JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN 
SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, 
MD, US, 

vol . 276, no. 10, 

9 March 2001 (2001-03-09), pages 

7346-7350, XP002248402 

ISSN: 0021-9258 

cited in the application 

page 7350 

page 7349 



-/-- 



1-21, 
47-56, 
60,111, 
112 



113-117 

24-26, 

32-35, 

38,40, 

47-56 , 

64-67, 

70-72, 



j )( I Further documents are listed in the continuation of box C. 



ID 



Patent family members are listed in annex. 



° Special categories of cited documents : 

•A" document defining the general state of the art which is not 
considered to be of particular relevance 

"E" earlier document but published on or after the international 
filing date 

"L* document which may throw doubts on priority claim(s) or 
which is cited to establish the publication date of another 
citation or other special reason (as specified) 

"O 1 document referring to an oral disclosure, use, exhibition or 
other means 

"P" document published prior to the international filing date but 
later than the priority date claimed 



*T later document published after the international filing date 
or priority date and not in conflict with the application but 
cited to understand the principle or theory underlying the 
invention 

"X" document of particular relevance; the claimed invention 
cannot be considered novel or cannot be considered to 
involve an inventive step when the document is taken alone 

"Y" document of particular relevance; the claimed invention 

cannot be considered to involve an Inventive step when the 
document is combined with one or more other such docu- 
ments, such combination being obvious to a person skilled 
in the art. 

document member of the same patent family 



Date of the actual completion of the international search 



8 June 2004 



Date of mailing of the international search report 



08. 07. 2004 



Name and mailing address of the ISA 

European Patent Office, P.B. 5818 Patentlaan 2 
NL - 2280 HV Rijswijk 
Tel. (+31-70) 340-2040, Tx. 31 651 epo nl, 
Fax: (+31-70) 340-3016 



Authorized officer 



Wagner, R 



Form PCT/lSA/210 (second sheet) (January 2004) 



INTERNATIONAL SEARCH REPORT 



Int >nal Application No 

PCT7GB 03/02804 



C.(Contlnuation) DOCUMENTS CONSIDERED TO BE RELEVANT 




wiiaUUil <JI UUUUintJlll, Willi IllUICdllun, wnere dp|-MUJ->i idle, ui nits leiyvaiu pciij^ciytiSj 


neievani to ciaim no. 






111,112 


Y 


page 7349 


27,28, 






36-38, 






zin 






47-56, 






64-67, 






70-72, 






111,112 




page 7349 


X 


WO 00/29004 A (PEPT0R LTD ;PLAKSIN DANIEL 


22,23, 




(ID) 25 May 2000 (2000-05-25) 


25, 






29-31, 












47-56, 






61-63, 






70-72, 






111,112 




example 2 




X 


REITER Y ET AL: "An antibody 


22 , 23 j 




single-domain phage display library of a 


25, 




native heavy chain variable region: 


29-31, 




isolation of functional single-domain VH 


38,40, 




molecules with a unique interface" 


47-56, 




UVJUl\IMr\L VJr rlULI-i'ULHIX DJ.ULUU I ? L-UIML/UN , UD, 


£1 — £^ 

O 1 UO y 




vol. 290, no. 3, 


70-72, 




1 January 1999 (1999-01-01), pages 


111,112 




685-698, XP004461990 




ISSN: 0022-2836 






page 687 




P,X 


W0 03/002609 A (MEDICAL RES COUNCIL 


41,42, 




;T0MLINS0N IAN (GB); WINTER GREG (GB); 


82,83 




IGNATOV) 9 January 2003 (2003-01-09) 




page 29, line 8-15 






t: A alllfJ 1 C U 




P,X 


W0 03/035694 A (MUYLDERMANS SERGE ; VLAAMS 


22,113 




INTERUNIV INST BIOTECH (BE)) 




1 May 2003 (2003-05-01) 






page 21; example 6 






Y 


W0 97/30084 A (GENETICS INST) 


24-26, 




21 August 1997 (1997-08-21) 


32-35, 






/in 






47-56, 






64-67, 






70-72, 






111,112 




example 6 




-/-- 





Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



INTERNATIONAL SEARCH REPORT 



lnt€ nal Application No 

PCT/GB 03/02804 



C.(Continuation) DOCUMENTS CONSIDERED TO BE RELEVANT 



Category 



Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



x 



EP 0 368 684 A (MEDICAL RES COUNCIL) 
16 May 1990 (1990-05-16) 

example 9 



example 5 



W0 98/40469 A (MCFARLAND CLIVE DAVID 
;UNDERW00D PATRICIA ANNE (AU); CARDIAC CRC 
N) 17 September 1998 (1998-09-17) 



page 6 

SMITH BRYAN J ET AL: "Prolonged in vivo 

residence times of antibody fragments 

associated with albumin" 

BI0C0NJUGATE CHEMISTRY, 

vol. 12, no. 5, September 2001 (2001-09), 

pages 750-756, XP002270731 

ISSN: 1043-1802 

the whole document 



VAN DEN BEUCKEN T ET AL: "Building novel 
binding ligands to B7.1 and B7.2 based on 
human antibody single variable light chain 
domains" 

JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, 
vol. 310, no. 3, 

13 July 2001 (2001-07-13), pages 591-601, 

XP004464206 

ISSN: 0022-2836 

abstract 

-/- 



39, 

47-56, 

111,112 

24-26, 

32-35, 

38,40, 

47-56, 

64-67, 

70-72, 

111,112 

27,28, 

36-38, 

40, 

47-56, 
64-67, 
70-72, 

l l 3* j l l 

27,28, 
36-38, 
40, 

47-56, 
64-67, 
70-72, 
111,112 



1-21, 
47-56, 
60,111, 
112 



1-21, 
47-56, 
60,111, 
112 



Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



INTERNATIONAL SEARCH REPORT 



Inte na! Application No 

PCT/GB 03/02804 



C.(Continuation) DOCUMENTS CONSIDERED TO BE RELEVANT 



Cateaorv ° 


Citation of document, with indication, where appropriate, of the relevant passages 


Relevant to claim No. 


A 


MUYLDERMANS S ET AL: "UNIQUE 


1-21, 




i ip h B j_ - am ^m b ■ a. *ai M 1 a a ft mmwm av a^ a ■ am ■ | v. _ | * m 1 M M ■ ^™ ftk ft ™ ^™ 

SINGLE-DOMAIN ANTIGEN BINDING FRAGMENTS 


M >>/ aa» _m 

47-56, 




•a* M a ft .tffete ft«a> am a a A ■ a pM | « am a V ■ % £ am am • % W m \ am *^ M ft ^% j^V H ft Ji 1 " 1 

DERIVES FROM NATURALLY OCCURRING CAMEL 


ft*** *X aft al al 

60,111, 




ft ft B|h a « 0 a m ■ ■ H M ■ ■ Ml M * aaa W rfh am. fta* f^*m « . 

HEAVY-CHAIN ANTIBODIES" 


a* vft a». 

112 




JOURNAL OF MOLECULAR RECOGNITION, HEYDEN & 

\m* *a# 1 \ 1 11 ft 1— \S | II J— la Vn* Ian / III 1 \ La* »a* • m am 1 ^a» 1 J * *~~ ■ m ^" 






SON LTD., LONDON, GB, 






vol. 12, no. 2, March 1999 (1999-03), 






4 ■ B ii i a Mm ate ate — * — aa. * * ate a. 

pages 131-140, XP009012180 






ISSN: 0952-3499 






abstract 




A 


RIECHMANN L ET AL- "Sinale domain 

1 \ •** la- V III If 11 11 1 La- La* I f \ Lm» * %y III \«J ■ V W l|| W*> 111 


1-21 

at Baa a> a 




antibodies: Comparison of camel VH and 


47-56, 




camel ised human VH domains" 


60,111, 




BIOSIS, 


112 




XP004187632 






the whole document 




A 


• * ^te. ft ft aa >te M ate m ^mm a M mm ate aar pa) a a >ft ar aaa 1 aa a f am »--te C*a> am f f a a 

HOLLIGER P ET AL: "RETARGETING SERUM 


1-21, 




aa a. a a a ft * a I ate. Jte 1 ate, k s ■ ■ aa> a ■ ft ft aa aaa ft ft aa aa ate. a, ^^a ate an ^^a aa ate. a aa a av ate av %aj Ma ^te^ a pj 

IMMUNOGLOBULIN WITH BISPECIFIC DIABODIES 


47-56, 




NATURE BIOTECHNOLOGY, NATURE PUBLISHING, 

1 V » 1 1 1 V k> La* aft* %a/ 1 La> 1 111 *a* La« W VI 1 4 1 H 1 • *a/ | \ kai | W a* haa aV >✓ 1 1 aV V * %al 4 


60, 111 . 




US, 


112 




a> _* an a _» am. ate aav / >^ ate a «a a. a# "V 

vol. 15, July 1997 (1997-07), pages 






632-636, XP002921893 






aa ate ate. a ■ - M ate ^av ate. * tew i _ 

ISSN: 1087-0156 






abstract 




A 


m m ate. as .ate a a ate aw a a a—_ am va^H a ■ vte teas a ft ate. afts am. ate ate. a 1 ft a a ■ a ■ aa* aaa 

US 5 644 034 A (RATHJEN DEBORAH ANN ET 


22,23, 




— ■ X a mmm mmm — ate. A .-^ ^- aate ate taaV a, _* X 

AL) 1 Ouly 1997 (1997-07-01) 


ate. aa> 

25, 




ate am ate —m 

29-31, 






38 40, 

\— ' ^ft* • 1 w * 






47-56, 






61-63, 






ftftf a- aaa ate 

70-72, 






111,112 




column 2 




■ 

A 


■ ft aV. ate. * f a— ate ate a^a a a V am ma> am, aaa va. am aaa fta ^^ft a ft ■ a • ~aaw m — a % ft ft a^M a "V 

W0 91/02078 A (PEPTIDE TECHNOLOGY LTD) 


22,23, 




am —m a— ■ ■ ^ft ate am. al / mm) ate a j a ate ate X 

21 February 1991 (1991-02-21) 


ate aa* 

25, ! 






29-31, 






38,40, 






47-56, 






61-63, 






70-72, 






111,112 




page 4, line 3,19 






-/~ 





Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



INTERNATIONAL SEARCH REPORT 



Ini onal Application No 

PCT/6B 03/02804 



C.(Continuation) DOCUMENTS CONSIDERED TO BE RELEVANT 



Category 



Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



A 



P,A 



TANHA J ET AL: "Optimal design features 
of camel ized human single-domain antibody 
1 ibraries . " 

THE JOURNAL OF BIOLOGICAL CHEMISTRY. 

UNITED STATES 6 JUL 2001, 

vol. 276, no. 27, 

6 July 2001 (2001-07-06), pages 

24774-24780, XP002283749 

ISSN: 0021-9258 

the whole document 

WO 02/072141 A (HERMAN WILLIAM) 
19 September 2002 (2002-09-19) 
the whole document 



1-117 



1-117 



Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



INTERNATIONAL SEARCH REPORT 



jrnational application No. 

PCT/GB 03/02804 



Box I Observations where certain claims were found unsearchable (Continuation of item 1 of first sheet) 

This International Search Report has not been established in respect of certain claims under Article 17(2)(a) for the following reasons: 
1. L Claims Nos.: 

because they relate to subject matter not required to be searched by this Authority, namely: 



!. I Claims Nos.: 

because they relate to parts of the International Application that do not comply with the prescribed requirements to such 
an extent that no meaningful International Search can be carried out, specif icaliy: 



3. I I Claims Nos.: 

because they are dependent claims and are not drafted in accordance with the second and third sentences of Rule 6.4(a). 

Box II Observations where unity of invention is lacking (Continuation of item 2 of first sheet) 

This International Searching Authority found multiple inventions in this international application, as follows: 

see additional sheet 



1 . | Y I As all required additional search fees were timely paid by the applicant, this International Search Report covers all 
L-^-J searchable claims. 



2. | | As all searchable claims could be searched without effort justifying an additional fee, this Authority did not invite payment 
of any additional fee. 



3. I I As only some of the required additional search fees were timely paid by the applicant, this International Search Report 
1 ' covers only those claims for which fees were paid, specifically claims Nos.: 



4. | | No required additional search fees were timely paid by the applicant. Consequently, this International Search Report is 
restricted to the invention first mentioned in the claims; it is covered by claims Nos.: 



Remark on Protest [ [ The additional search fees were accompanied by the applicant's protest. 

[ X | No protest accompanied the payment of additional search fees. 



Form PCT/ISA/210 (continuation of first sheet (1)) (July 1998) 



International Application No. PCT/ GB 03/02804 



FURTHER INFORMATION CONTINUED FROM PCT/ISA/ 210 



This International Searching Authority found multiple (groups of) 
inventions in this international application, as follows: 

1. Claims: 1-7,8-10,11-21,47-56 (all 10 in part), 

60 (in part), 111-112 (both in part) 



Ligand comprising a first immunoglobulin variable domain 
having a first antigen or epitope specificity and a second 
immunoglobulin variable domain having a second antigen or 
epitope specificity wherein one or both of the antigens or 
epitopes acts to increase the half-life of the ligand in 
vivo, and the variable domains are not complementary to one 
another. 



2. Claims: 22,23,25,29-31, 38 (in part), 40, 

47-56 (all 10 part), 61-63, 70-72 (all 3 in part), 
111-112 (both in part) 



Single domain antibody monomer ligand specific for TNF- . 



3. Claims: 24,26,25,32-35, 38 (in part), 40, 

47-56 (all 10 in part), 64-67, 
70-72 (all 3 in part), 111-112 (both in part) 



Single domain antibody monomer ligand specific for TNF 
receptor 1. 



4. Claims: 27, 28, 36,37, 38 (in part)40, 

47-56 (all 10 in part), 64-67, 
70-72 (all 3 in part), 111-112 (both in part) 



Single domain antibody monomer ligand specific for serum 
albumin. 



5. Claims: 39, 47-56 (all 10 in part), 

111-112 (both in part). 



Single domain antibody monomer ligand comprising a terminal 
Cys residue. 



6. Claims: 41-46, 47-56 (all 10 in part), 60 (in part). 

A dual specific ligand comprising at least a single domain 
antibody monomer according to inventions 2-5. 



page 1 of 2 



International Application No. PC77 GB 03/02804 



FURTHER INFORMATION CONTINUED FROM PCT/ISA/ 210 



7. Claims: 73-81, 82-104, 105-110 

A closed conformation multi specif ic ligand comprising a 
first epitope binding domain having a first epitope binding 
specificity and a non-complementary second epitope binding 
domain having a second epitope binding specificity, wherein 
the first and second binding specificities are capable of 
competing for epitope binding such that the closed 
conformation multi-specific ligand cannot bind both 
epitopes simultaneously. Methods for producing the closed 
conformation multispecif ic ligand. Methods using the closed 
conformation multispecif ic ligands. 



8. Claims: 113-117 

Method for preparing a chelating multimeric ligand, specific 
for adjacent epitopes. 



page 2 of 2 



INTERNATIONAL SEARCH REPORT 



In ional Application No 

PCT/GB 03/02804 



Patent document 


Publication 




Patent family 


Publication 


cited in search report 


date 




member(s) 


date 


WO 0029004 A 


25-05-2000 


Al 1 
MU 


/OOcUl DC 


11-09-2003 




Al 1 
nil 


04o0yyy M 


U5-06-2000 




CA 


2351669 Al 


25-05-2000 




EP 


1131079 Al 


12-09-2001 




Lin 

WU 


nnoonn/i ai 
uu<iyuu4 Ml 


ocr a TT OAAA 

2b-05-2000 




IV L 


O11400 M 


on n a oaao 

29-04-2003 




us 


2002012909 Al 


31-01-2002 


W0 03002609 A 


09-01-2003 


r a 

LM 


£44/obl Ml 


An ai or\ no 

U9-01-2003 




EP 


1399484 A2 


24-03-2004 




WO 


03002609 A2 


09-01-2003 


W0 03035694 A 


01-05-2003 


t. r 


loUDooD Ml 


no act onno 




wu 


no.f)OK£Q/l A9 

uouo£>oy4 t\c 


ni nzr onno 
Ui-Ub-ZUOo 


W0 9730084 A 


21-08-1997 


1 IQ 
Uo 


Do4o0/o M 


ni *io mno 

1)1-1^-1998 




1 IQ 
Uo 


o/l/iool A 


o "V r\i iaao 

2/-01-1998 




AU 


2268697 A 


02-09-1997 




WO 


9730084 Al 


21-08-1997 




1 IQ 
Uo 


ooccy i c d! 


2/-11-2001 




lie 
uo 


CQQ1£7E A 
OOi7lD/D M 


n^ n / i nnn 

uo-U4-l999 




1 IQ 
UO 


9nnoiAyi7iA at 

CUUC.IOH / 10 Ml 


n*7 i i onno 

U/-11-Z002 


EP 0368684 A 


16-05-1990 


AT 
M 1 


i noAo. 1 t 

lU*lDol I 


i c no 1 nn/i 
lb-Uo-1994 




Al 1 
HU 


f\1A 1 OA DO 
0o41o0 DC 


i o no i nno 

lo-U<£~199o 




Al 1 
MU 


yiFoni qq a 


oo nn i nnn 
<io-Ub-1990 




PA 
UM 


cVKJcoOa Ml 


ii nc 1 nnn 

1 l-ub-1990 




UL 


OorloODo Ul 


1 A r\A i nn a 
14— U4— 1994 






AQOI OAKQ TO 

Ooyioooo 


no nn i nn/i 
Uo-09-1994 




UN 


1A/17QH A 

104/yu M 


n*7 nn i nnn 
U/-U9-1990 




FP 

tr 


UoOoOoH Ml 


i ^ nc i nnn 

io-Ub-1990 




FQ 
to 


onnono7 to 
cUQcVc/ 1 o 


n i n*7 i nn /i 
Ui— U/— 1994 




un 

WU 


cinnc i /i a a i 
yUUb!44 Ml 


1 i nc i nnn 

1/-05-1990 




ip 


OQIQQQn DO 

^yiyoyu dc 


1 n n"7 i nnn 

19-U/-1999 




ur 


oDUfLoUl I 


ot n^r mni 

£/-0o-1991 




Nl\ 


lo4oOU Dl 


ai r\ A "1 A A A 

01-04-1999 




NO 


903059 A 


07-09-1990 




US 


2003130496 Al 


10-07-2003 




1 IQ 

Uo 


*iUUoll40oy Ml 


19-06-2003 




us 


6248516 Bl 


19-06-2001 




us 


6545142 Bl 


08-04-2003 


W0 9840469 A 


17-09-1998 


All 
MU 


oou<i/yo m 


on nn i nno 

£9— 09— 1998 




WU 


yo4U4oy mi 


i "7 nn i nno 

1/-U9-1998 


US 5644034 A 


01-07-1997 


1 IQ 
Uo 


onnoi 0QC77 ai 
tuuoidyo// mi 


o/i H7 onno 




1 IQ 
Uo 


onnoi oqcqh ai 

^.UUoloyooU Mi 


o^ n~7 onno 
ch-[j / - CUV 6 




1 IQ 
UO 


OftfiO.1 £OQ/1 Q Al 
<LUUolO£y4o Ml 


oo no onno 

Zo-Uo-2003 




MQ 
UO 


onno,*? QcnoQ ai 
^uuoiooUiiy mi 


1 7 H7 onno 

1/-U/-2003 




I IQ 
Uo 


onnoiQOA7o ai 
^UUoiyyo/o Al 


OO 1 A OAAO 

23-10-2003 




1 IQ 

Uo 


onn*3000Q7n ai 

^uuorfiovy/u mi 


1 O 1 O OAAO 

18-12-2003 




1 IQ 
Uo 


onnoi 7non/i ai 
cUUoi/UcVh Ml 


ii nn oaao 

11-U9-2003 




1 IQ 
Uo 


onnoonon/io ai 
£UUo£UoU4y Ml 


f\£l 1 1 OAAO 

06-11-2003 




us 


2004002588 Al 


01-01-2004 




us 


2003171553 Al 


11-09-2003 




us 


2003166874 Al 


04-09-2003 




us 


2003216552 Al 


20-11-2003 




us 


2004002589 Al 


01-01-2004 



Form PCT/ISA/210 {patent family annex) (January 2004) 



INTERNATIONAL SEARCH REPORT 



PCT/GB 03/02804 



Patent document 
cited in search report 


Publication 
date 


Patent family 
member(s) 


Publication 
date 


US 5644034 A 




us 


2003208047 


Al 


UU 1 l*"6UUo 




us 


2003171554 


Al 


1 1 _ oo— 900? 

J. JL U7 £UUO 




us 


2003171555 


Al 


1 i ~nQ— 900? 

1 J. U7 lUUO 




us 


2004002590 


Al 


01 —01 -9004 

ul UX LUUH 




us 


2004092721 


Al 


l Q— OR— 9004 

XO UO tUU't 




us 


2003225254 


Al 


04-1 9-900? 

UH 1C LuWO 




us 


6416757 


Bl 


00—07-9009 

U7 U / luUc 




us 


2003232971 


Al 


ia-i 9-900? 




us 


6593458 


Bl 


i R-07-900? 

X O U / C-UUO 




us 


2001018507 


Al 


?o~oa-90oi 

OU UO cuul 




us 


2001023287 


Al 


90-00-9001 

£.U U7 Luul 




us 


2001018508 


Al 


?o-oa-9noi 

OU UO LvJU X 




us 


5959087 


A 


po_00-1 QQQ 

C_ O U7 X777 




AU 


640400 


B2 


96-08-100? 

C.U uo x77o 




AU 


6145490 


A 


i i-o?-1901 

4.x uo x -/ 7 i 




wo 


9102078 


Al 


pi —09-1 QQ1 




CA 


2064915 


Al 


Oft-09-1001 

UO U£_ X77 X 




DE 


69027121 


Dl 

t-J J- 


97-06-1 OOfi 

C / UU X77U 




DE 


69027121 


T2 


1 4-1 1 -1006 

14 XX X77U 




DK 


486526 


T3 


24-06-1996 




EP 


0486526 


Al 


27-05-1992 




JP 


3443119 


B2 






OP 


2003096096 


A 


o?-04-90o? 

UO U *t t-UUJ 




OP 


2003231699 


A 


1 0— Oft-900? 

X 27 UO cUUJ 


WO 9102078 A 


21-02-1991 


AU 


640400 


B2 


9fi-0R~1 QQ? 

L\\J UO X7TO 




AU 


6145490 


A 


i i -0?-1QQ1 

XX UO 157 X 




WO 


9102078 


Al 


91 -09-1 001 

c. X U£. 177 X 




EP 


0486526 


Al 


P7— OR-1 OOP 

£./ UO X 7 7 £. 




CA 


2064915 


Al 


0£-09-iooi 

UO U£ X77X 




DE 


69027121 


Dl 


97-06-1006 




DE 


69027121 


T2 


14-1 1-1006 

in XX 177U 




DK 


486526 


T3 


94-06-1006 

UU I7-7U 




JP 


3443119 


B2 


09-00-900? 

U£_ U7 lUUJ 




JP 


2003096096 


A 


o?-o4-90o? 

UO U*T C.UUO 




JP 


2003231699 


A 


1 0-08-900? 

17 UO LuUJ 




US 


2003162948 


Al 


p«-oa~90o? 




US 


2003232970 


Al 


1 Q-i 9-900? 

XO X C LUUJ 




US 


2004002588 


Al 


01-01 -9004 

UX UX LUuH 




US 


2003171553 


Al 


i i ~oo-90o? 

X X w7 lUUJ 




US 


2003166874 


Al 


04-00-900? 

U*t U7 lUUJ 




US 


2003216552 


Al 


po-1 1-900? 

C-U X X L.UU o 




US 


2004002589 


Al 


U X U X (CUUH 1 




US 


2003208047 


Al 


06-1 1 -900? 

Uv XX C\J w O 




US 


5644034 


A 


01-07-1007 

UX U / 177/ 




US 


2003171554 


Al 


1 1-00-900? 

XX U 7 t_U U O 




US 


2003171555 


Al 


1 1 -00-900? 

XX U7 cUuJ 




US 


2004002590 


Al 


01 —01 -9004 

UX UX LUUH 




US 


2004092721 


Al 


1 ?-05-9004 

XO UO lUUH 




US 


2003225254 


Al 

r\ X 


04—1 9-900? 




us 


6416757 


Bl 


00—07-9009 

u7 U/ C.UUC 




us 


2003232971 


Al 


18-12-2003 




us 


6593458 


Bl 


15-07-2003 




us 


2001018507 


Al 


30-08-2001 




us 


2001023287 


Al 


cxj— uy— £uui 




us 


2001018508 


Al 


30-08-2001 


W0 02072141 A 


19-09-2002 


wo 


02072141 


A2 


19-09-2002 



Form PCT/tSA/210 (patent family annex) (January 2004) 



INTERNATIONAL SEARCH REPORT 


li tional Application No 

PCT/GB 03/02804 


Patent document 
cited in search report 


Publication 
date 


Patent family 
member(s) 


Publication 
date 



WO 02072141 A W0 03057732 A2 17-07-2003 



, , . I 

Form PCT/iSA/210 (patent family annex) (January 2004)